1
|
Yu K, Estonian Biobank Research Team, Estrada K, Esko T, Kals M, Nikopensius T, Kronberg J, Võsa U, Wuster A, Bomba L. Plasma Metabolic Outliers Identified in Estonian Human Knockouts. Metabolites 2025; 15:323. [PMID: 40422899 DOI: 10.3390/metabo15050323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Background/Objectives: Metabolomics, in combination with genetic data, is a powerful approach to study the biochemical consequences of genetic variation. We assessed the impact of human gene knockouts (KOs) on the metabolite levels of Estonia Biobank (EstBB) participants and integrated the results with electronic health record data. Methods: In 150,000 EstBB genotyped participants, we identified 723 KOs with 152 different predicted loss of function (pLoF) variants in 115 genes. For those KOs and 258 controls, 1387 metabolites were profiled using ultra-high-performance liquid chromatography-tandem mass spectrometry. Results: We identified 48 associations linking rare pLoF variants in 22 genes to 43 metabolites. Out of 48 associations, 27 (56%) were found in genes that cause inborn errors of metabolism. The top associations identified in our analysis included genes and metabolites involved in the degradation pathway of the pyrimidine bases uracil and thymine (DPYD and UPB1). We found DPYD gene KOs to be associated with elevated levels of Uracil, confirming that DPD-deficiency is a leading cause of severe 5-Fluorouracil toxicity. Overall, 54% of reported associations are gene targets of approved drugs or bioactive drug-like compounds. Conclusions: Our findings contribute to assessing the impact of human KOs on metabolite levels and offer insights into gene functions, disease mechanism, and drug target validation.
Collapse
Affiliation(s)
- Ketian Yu
- Genomics, BioMarin Pharmaceutical, Novato, CA 94949, USA
| | | | - Karol Estrada
- Genomics, BioMarin Pharmaceutical, Novato, CA 94949, USA
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Mart Kals
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Tiit Nikopensius
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Arthur Wuster
- Genomics, BioMarin Pharmaceutical, Novato, CA 94949, USA
| | - Lorenzo Bomba
- Genomics, BioMarin Pharmaceutical, Novato, CA 94949, USA
| |
Collapse
|
2
|
Tain YL, Hsu CN. The NOS/NO System in Renal Programming and Reprogramming. Antioxidants (Basel) 2023; 12:1629. [PMID: 37627624 PMCID: PMC10451971 DOI: 10.3390/antiox12081629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Nitric oxide (NO) is a gaseous signaling molecule with renoprotective properties. NO can be produced in NO synthase (NOS)-dependent or -independent manners. NO deficiency plays a decisive role in chronic kidney disease (CKD). Kidney development can be affected in response to adverse intrauterine conditions that induce renal programming, thereby raising the risk of developing CKD in adulthood. Conversely, detrimental programming processes could be postponed or halted prior to the onset of CKD by early treatments, namely reprogramming. The current review provides an overview of the NOS/NO research performed in the context of renal programming and reprogramming. NO deficiency has been increasingly found to interact with the different mechanisms behind renal programming, such as oxidative stress, aberrant function of the renin-angiotensin system, disturbed nutrient-sensing mechanisms, dysregulated hydrogen sulfide signaling, and gut microbiota dysbiosis. The supplementation of NOS substrates, the inhibition of asymmetric dimethylarginine (ADMA), the administration of NO donors, and the enhancement of NOS during gestation and lactation have shown beneficial effects against renal programming in preclinical studies. Although human data on maternal NO deficiency and offspring kidney disease are scarce, experimental data indicate that targeting NO could be a promising reprogramming strategy in the setting of renal programming.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
3
|
Rodionov RN, Jarzebska N, Burdin D, Todorov V, Martens-Lobenhoffer J, Hofmann A, Kolouschek A, Cordasic N, Jacobi J, Rubets E, Morawietz H, O'Sullivan JF, Markov AG, Bornstein SR, Hilgers K, Maas R, Pfluecke C, Chen Y, Bode-Böger SM, Hugo CPM, Hohenstein B, Weiss N. Overexpression of alanine-glyoxylate aminotransferase 2 protects from asymmetric dimethylarginine-induced endothelial dysfunction and aortic remodeling. Sci Rep 2022; 12:9381. [PMID: 35672381 PMCID: PMC9174227 DOI: 10.1038/s41598-022-13169-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/20/2022] [Indexed: 12/03/2022] Open
Abstract
Elevated plasma concentrations of asymmetric dimethylarginine (ADMA) are associated with an increased risk of mortality and adverse cardiovascular outcomes. ADMA can be metabolized by dimethylarginine dimethylaminohydrolases (DDAHs) and by alanine-glyoxylate aminotransferase 2 (AGXT2). Deletion of DDAH1 in mice leads to elevation of ADMA in plasma and increase in blood pressure, while overexpression of human DDAH1 is associated with a lower plasma ADMA concentration and protective cardiovascular effects. The possible role of alternative metabolism of ADMA by AGXT2 remains to be elucidated. The goal of the current study was to test the hypothesis that transgenic overexpression of AGXT2 leads to lowering of plasma levels of ADMA and protection from vascular damage in the setting of DDAH1 deficiency. We generated transgenic mice (TG) with ubiquitous overexpression of AGXT2. qPCR and Western Blot confirmed the expression of the transgene. Systemic ADMA levels were decreased by 15% in TG mice. In comparison with wild type animals plasma levels of asymmetric dimethylguanidino valeric acid (ADGV), the AGXT2 associated metabolite of ADMA, were six times higher. We crossed AGXT2 TG mice with DDAH1 knockout mice and observed that upregulation of AGXT2 lowers plasma ADMA and pulse pressure and protects the mice from endothelial dysfunction and adverse aortic remodeling. Upregulation of AGXT2 led to lowering of ADMA levels and protection from ADMA-induced vascular damage in the setting of DDAH1 deficiency. This is especially important, because all the efforts to develop pharmacological ADMA-lowering interventions by means of upregulation of DDAHs have been unsuccessful.
Collapse
Affiliation(s)
- Roman N Rodionov
- University Center for Vascular Medicine and Department of Medicine III, Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| | - Natalia Jarzebska
- University Center for Vascular Medicine and Department of Medicine III, Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- Department of Anesthesiology and Critical Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Dmitrii Burdin
- Department of General Physiology, Saint-Petersburg State University, 199034, Saint-Petersburg, Russia
| | - Vladimir Todorov
- Department of Medicine III, Section Nephrology, Technische Universität Dresden, 01307, Dresden, Germany
| | | | - Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Anne Kolouschek
- University Center for Vascular Medicine and Department of Medicine III, Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Johannes Jacobi
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Elena Rubets
- Department of General Physiology, Saint-Petersburg State University, 199034, Saint-Petersburg, Russia
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - John F O'Sullivan
- The University of Sydney, Charles Perkins Centre, Sydney, NSW, Australia
- The University of Sydney, Heart Research Institute, Sydney, NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Alexander G Markov
- Department of General Physiology, Saint-Petersburg State University, 199034, Saint-Petersburg, Russia
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Karl Hilgers
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Christian Pfluecke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden, 01307, Dresden, Germany
| | - YingJie Chen
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, 5455, USA
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-Von-Guericke University, 39120, Magdeburg, Germany
| | - Christian P M Hugo
- Department of Medicine III, Section Nephrology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Bernd Hohenstein
- Department of Medicine III, Section Nephrology, Technische Universität Dresden, 01307, Dresden, Germany
| | - Norbert Weiss
- University Center for Vascular Medicine and Department of Medicine III, Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| |
Collapse
|
4
|
Hall JA, Panickar KS, Brockman JA, Jewell DE. Cats with Genetic Variants of AGXT2 Respond Differently to a Dietary Intervention Known to Reduce the Risk of Calcium Oxalate Stone Formation. Genes (Basel) 2022; 13:791. [PMID: 35627178 PMCID: PMC9141165 DOI: 10.3390/genes13050791] [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: 03/19/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
This study was completed to evaluate a genotype-specific nutritional intervention for reducing the risk of calcium oxalate stone formation. Serum metabolomic profiles and genotypes of 445 cats in the colony at Hill’s Pet Nutrition, Inc (Topeka, KS, USA)were assessed in a genome-wide association study, and revealed an association between genetic variants of alanine-glyoxylate aminotransferase 2 (AGXT2) and 2-oxoarginine. The most significant single nucleotide polymorphisms (SNP) associated with 2-oxoarginine was at position chrA1:212069607, [G/A] (p < 3.687 × 10−17). This SNP explained approximately 15% of the variance in 2-oxoarginine concentrations. The distribution of genotype frequencies was 0.07 AA, 0.39 AG, and 0.54 GG, with a mean relative 2-oxoarginine concentration for each genotype of 0.45 AA, 0.92 AG, and 1.27 GG, indicating a subtractive effect of the minor allele (A). Serum concentrations of two AGXT2 substrates, symmetric/asymmetric dimethylarginines (SDMA/ADMA) and β-aminoisobutyrate (BAIB) were also strongly associated with SNP chrA1:212069607 (p < 1.43 × 10−12 and p < 2.30 × 10−14, respectively). These two AGXT2 substrates were increased with the minor allele (A), indicating that the variant of the AGXT2 gene results in decreased aminotransferase activity. Additionally, the lifetime history of stone incidence showed that cats with the AA variant of AGXT2 SNP had a 2.515× increased incidence of stones compared with cats having the GG variant (p = 0.019). In a subsequent study assessing AGXT2 genotypes, cats (n = 10 GG, 4 AG, 9 AA) were fed control or test food (containing betaine at 0.500%, and the botanicals green tea, fenugreek and tulsi at 0.25, 0.025, and 0.0015%, respectively) in a cross-over study design. Stone risk analysis was conducted on urine samples after feeding control or test food for 28 days each. A calcium oxalate titration test (COT) was performed to assess the amount of added Ox−2 (per L) required to initiate calcium oxalate crystal formation. Cats with the GG variant of the AGXT2 SNP required more added oxalate to initiate urine crystal formation after consuming test food compared with control food, indicating a decreased risk of oxalate crystal formation in GG cats. In addition, urine oxalate concentrations showed an overall effect of test food independent of genotype (p = 0.0009), which resulted in lower oxalate concentrations after consuming test food compared with control food. These data indicate that cats with the GG-specific variant of AGXT2 should benefit from a reduced risk of calcium oxalate stone formation after consuming a betaine and botanical dietary enhancement.
Collapse
Affiliation(s)
- Jean A. Hall
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Kiran S. Panickar
- Science & Technology Center, Hill′s Pet Nutrition, Inc., Topeka, KS 66617, USA; (K.S.P.); (J.A.B.)
| | - Jeffrey A. Brockman
- Science & Technology Center, Hill′s Pet Nutrition, Inc., Topeka, KS 66617, USA; (K.S.P.); (J.A.B.)
| | - Dennis E. Jewell
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA;
| |
Collapse
|
5
|
ADMA and homoarginine independently predict mortality in critically ill patients. Nitric Oxide 2022; 122-123:47-53. [DOI: 10.1016/j.niox.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/20/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022]
|
6
|
Hannemann J, Zummack J, Hillig J, Rendant-Gantzberg L, Böger R. Association of Variability in the DDAH1, DDAH2, AGXT2 and PRMT1 Genes with Circulating ADMA Concentration in Human Whole Blood. J Clin Med 2022; 11:jcm11040941. [PMID: 35207213 PMCID: PMC8877358 DOI: 10.3390/jcm11040941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/10/2022] Open
Abstract
Asymmetric dimethylarginine is an endogenous inhibitor of nitric oxide synthesis and a cardiovascular risk factor. Its regulation has been studied extensively in experimental models, but less in humans. We studied common single-nucleotide polymorphisms (SNPs) in genes encoding for enzymes involved in ADMA biosynthesis and metabolism, i.e., PRMT1, DDAH1, DDAH2, and AGXT2, and assessed their associations with blood ADMA concentration in 377 unselected humans. The minor allele of DDAH1 SNP rs233112 was significantly more frequent in individuals with ADMA in the highest tertile or in the highest quartile, as was the major allele of DDAH2 rs805304. A combined genotype comprising both SNPs showed a significant genotype–phenotype association, with increasing ADMA concentration by an increasing number of inactive alleles. SNPs in the AGXT2 and PRMT1 genes showed no significant associations with blood ADMA concentration. Our study provides comprehensive evidence that DDAH1 and DDAH2 are the major enzymes regulating blood ADMA concentration, whilst PRMT1 indirectly affects ADMA, and AGXT2 may act as a back-up enzyme in ADMA metabolism under pathophysiological conditions only.
Collapse
|
7
|
Bariatric Surgery Improves the Atherogenic Profile of Circulating Methylarginines in Obese Patients: Results from a Pilot Study. Metabolites 2021; 11:metabo11110759. [PMID: 34822417 PMCID: PMC8624057 DOI: 10.3390/metabo11110759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 12/04/2022] Open
Abstract
Bariatric surgery improves obesity-related comorbidities. Methylarginines are biomarkers of cardiometabolic risk, liver steatosis, and insulin resistance. Here, we aimed to investigate methylarginines in obese patients undergoing bariatric surgery and compared them to age- and sex-matched healthy subjects. Thirty-one obese patients who underwent bariatric surgery and 31 healthy individuals were used for this retrospective study. The basal serum methylarginine levels were determined in the healthy individuals and the obese patients, before surgery and 6 and 12 months after surgery, by mass spectrometry. Compared with the healthy individuals, the obese patients displayed elevated monomethylarginine (mean change: +95%, p < 0.001), asymmetric-dimethylarginine (+105%, p < 0.001), symmetric-dimethylarginine (+25%, p = 0.003), and dimethylguanidino valerate (+32%, p = 0.008) concentrations. Bariatric surgery durably reduced the body mass index by 28% (12 months, 95%CI: 24–33, p = 0.002) and improved plasma lipids, insulin resistance, and liver function. Bariatric surgery reduced the serum levels of monomethylarginine and asymmetric-dimethylarginine by 12% (95%CI: 6–17) and 36% (95%CI: 27–45) (12 months, p = 0.003), respectively, but not symmetric-dimethylarginine or dimethylguanidino valerate. The monomethylarginine and asymmetric-dimethylarginine concentrations were strongly correlated with markers of dyslipidemia, insulin resistance, and a fatty liver. Serum dimethylguanidino valerate was primarily correlated with glycemia and renal function, whereas serum symmetric-dimethylarginine was almost exclusively associated with renal function. In conclusion, the monomethylarginine and asymmetric-dimethylarginine levels are efficiently decreased by bariatric surgery, leading to a reduced atherogenic profile in obese patients. Methylarginines follow different metabolic patterns, which could help for the stratification of cardiometabolic disorders in obese patients.
Collapse
|
8
|
Kopaliani I, Jarzebska N, Billoff S, Kolouschek A, Martens-Lobenhoffer J, Bornstein SR, Bode-Böger SM, Ragavan VN, Weiss N, Mangoni AA, Deussen A, Rodionov RN. Overexpression of dimethylarginine dimethylaminohydrolase 1 protects from angiotensin II-induced cardiac hypertrophy and vascular remodeling. Am J Physiol Heart Circ Physiol 2021; 321:H825-H838. [PMID: 34533401 DOI: 10.1152/ajpheart.00064.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
Cardiovascular complications are the leading cause of death, and elevated levels of asymmetric dimethyarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, are implicated in their pathophysiology. We investigated the role of dimethylarginine dimethylaminohydrolase 1 (DDAH1), an enzyme hydrolyzing ADMA, in prevention of cardiovascular remodeling during hypertension. We hypothesized that the animals overexpressing DDAH1 will be protected from angiotensin II (ANG II)-induced end organ damage. Angiotensin II (ANG II) was infused in two doses: 0.75 and 1.5 mg/kg/day in DDAH1 transgenic mice (DDAH1 TG) and wild-type (WT) littermates for 2 or 4 wk. Echocardiography was performed in the first and fourth weeks of the infusion, systolic blood pressure (SBP) was measured weekly, and cardiac hypertrophy and vascular remodeling was assessed by histology. Increase in SBP after 1 wk of ANG II infusion was not different between the groups, whereas TG mice had lower SBP at later time points. TG mice were protected from cardiovascular remodeling after 2 wk of ANG II infusion in the high dose and after 4 wk in the moderate dose. TG mice had higher left ventricular lumen-to-wall ratio, lower cardiomyocyte cross-sectional area, and less interstitial fibrosis compared with WT controls. In aorta, TG mice had less adventitial fibrosis, lower medial thickness with preserved elastin content, lower counts of inflammatory cells, lower levels of active matrix metalloproteinase-2, and showed better endothelium-dependent relaxation. We demonstrated that overexpression of DDAH1 protects from ANG II-induced cardiovascular remodeling and progression of hypertension by preserving endothelial function and limiting inflammation.NEW & NOTEWORTHY We showed that overexpression of dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects from angiotensin II-induced cardiovascular damage, progression of hypertension, and adverse vascular remodeling in vivo. This protective effect is associated with decreased levels of asymmetric dimethylarginine, preservation of endothelial function, inhibition of cardiovascular inflammation, and lower activity of matrix metalloproteinase-2. Our findings are highly clinically relevant, because they suggest that upregulation of DDAH1 might be a promising therapeutic approach against angiotensin II-induced end organ damage.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Department of Physiology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Natalia Jarzebska
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Dresden University of Technology, Dresden, Germany
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Silke Billoff
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
| | - Anne Kolouschek
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
| | | | - Stefan R Bornstein
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| | - Vinitha N Ragavan
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Norbert Weiss
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Andreas Deussen
- Department of Physiology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Roman N Rodionov
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
9
|
Wali JA, Koay YC, Chami J, Wood C, Corcilius L, Payne RJ, Rodionov RN, Birkenfeld AL, Samocha-Bonet D, Simpson SJ, O'Sullivan JF. Nutritional and metabolic regulation of the metabolite dimethylguanidino valeric acid: an early marker of cardiometabolic disease. Am J Physiol Endocrinol Metab 2020; 319:E509-E518. [PMID: 32663097 PMCID: PMC7509244 DOI: 10.1152/ajpendo.00207.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dimethylguanidino valeric acid (DMGV) is a marker of fatty liver disease, incident coronary artery disease, cardiovascular mortality, and incident diabetes. Recently, it was reported that circulating DMGV levels correlated positively with consumption of sugary beverages and negatively with intake of fruits and vegetables in three Swedish community-based cohorts. Here, we validate these results in the Framingham Heart Study Third Generation Cohort. Furthermore, in mice, diets rich in sucrose or fat significantly increased plasma DMGV concentrations. DMGV is the product of metabolism of asymmetric dimethylarginine (ADMA) by the hepatic enzyme AGXT2. ADMA can also be metabolized to citrulline by the cytoplasmic enzyme DDAH1. We report that a high-sucrose diet induced conversion of ADMA exclusively into DMGV (supporting the relationship with sugary beverage intake in humans), while a high-fat diet promoted conversion of ADMA to both DMGV and citrulline. On the contrary, replacing dietary native starch with high-fiber-resistant starch increased ADMA concentrations and induced its conversion to citrulline, without altering DMGV concentrations. In a cohort of obese nondiabetic adults, circulating DMGV concentrations increased and ADMA levels decreased in those with either liver or muscle insulin resistance. This was similar to changes in DMGV and ADMA concentrations found in mice fed a high-sucrose diet. Sucrose is a disaccharide of glucose and fructose. Compared with glucose, incubation of hepatocytes with fructose significantly increased DMGV production. Overall, we provide a comprehensive picture of the dietary determinants of DMGV levels and association with insulin resistance.
Collapse
Affiliation(s)
- Jibran A Wali
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Yen Chin Koay
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, School of Medicine, The University of Sydney, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Jason Chami
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, School of Medicine, The University of Sydney, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Courtney Wood
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, School of Medicine, The University of Sydney, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Leo Corcilius
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Roman N Rodionov
- University Center for Vascular Medicine and Department of Medicine III-Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas L Birkenfeld
- Department of Internal Medicine, Division of Endocrinology, Diabetology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Tübingen, Germany
| | - Dorit Samocha-Bonet
- The Garvan Institute of Medical Research, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Science, School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - John F O'Sullivan
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, School of Medicine, The University of Sydney, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Jarzebska N, Georgi S, Jabs N, Brilloff S, Maas R, Rodionov RN, Zietz C, Montresor S, Hohenstein B, Weiss N. Kidney and liver are the main organs of expression of a key metabolic enzyme alanine:glyoxylate aminotransferase 2 in humans. ATHEROSCLEROSIS SUPP 2020; 40:106-112. [PMID: 31818439 DOI: 10.1016/j.atherosclerosissup.2019.08.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The metabolic syndrome is a cluster of cardiovascular risk factors and is highly predictive for development of cardiovascular diseases. An association between elevated plasma levels of the endogenous inhibitor of nitric oxide synthases asymmetric dimethylarginine (ADMA) and risk of cardiovascular diseases has been demonstrated in numerous epidemiological studies. ADMA can be catabolized by dimethylarginine dimethylaminohydrolase (DDAH) or metabolized through a much less understood alternative pathway by alanine:glyoxylate aminotransferase 2 (AGXT2) with the formation of α-keto-δ-(N,N-dimethylguanidino)valeric acid (ADGV). Previous RT-PCR and Western Blot studies suggested that Agxt2 is expressed in the mouse kidney and liver at comparable levels, while Northern Blot and in-situ RNA-hybridisation experiments demonstrated that the kidney is the main organ of Agxt2 expression in rats. Given this discrepancy, the goal of the current study was to analyse the expression of AGXT2 in human tissues. MATERIAL AND METHODS We analyzed AGXT2 expression in human tissues from a normal tissue bank by RT-PCR and further validated the results by Western Blot. We also performed immunohistochemical staining for AGXT2 and double fluorescent staining with an anti-AGXT2 antibody and a monoclonal anti-mitochondrial antibody. RESULTS We saw the strongest expression of AGXT2 in the kidney and liver and confirmed this results on protein level. By IHC staining we were able to show that AGXT2 is present in the convoluted tubule in the kidney and in the liver hepatocytes. The double fluorescent staining revealed mitochondrial localization of AGXT2. CONCLUSIONS Our current data suggest that both hepatocytes and kidney tubular epithelial cells are the major sources of AGXT2 in humans. We also demonstrated the mitochondrial localization of human AGXT2 enzyme.
Collapse
Affiliation(s)
- Natalia Jarzebska
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Sophia Georgi
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Normund Jabs
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Silke Brilloff
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roman N Rodionov
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Christian Zietz
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sabrina Montresor
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bernd Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Norbert Weiss
- University Center for Vascular Medicine & Department of Medicine III - Section Angiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
11
|
Stautemas J, Van Kuilenburg ABP, Stroomer L, Vaz F, Blancquaert L, Lefevere FBD, Everaert I, Derave W. Acute Aerobic Exercise Leads to Increased Plasma Levels of R- and S-β-Aminoisobutyric Acid in Humans. Front Physiol 2019; 10:1240. [PMID: 31611815 PMCID: PMC6773837 DOI: 10.3389/fphys.2019.01240] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/10/2019] [Indexed: 12/29/2022] Open
Abstract
Recently, it was suggested that β-aminoisobutyric acid (BAIBA) is a myokine involved in browning of fat. However, there is no evidence for an acute effect of exercise supporting this statement and the metabolic distinct enantiomers of BAIBA were not taken into account. Concerning these enantiomers, there is at this point no consensus about resting concentrations of plasma R- and S-BAIBA. Additionally, a polymorphism of the alanine - glyoxylate aminotransferase 2 (AGXT2) gene (rs37369) is known to have a high impact on baseline levels of total BAIBA, but the effect on the enantiomers is unknown. Fifteen healthy recreationally active subjects, with different genotypes of rs37369, participated in a randomized crossover trial where they exercised for 1 h at 40% of Ppeak or remained at rest. Plasma samples were analyzed for R- and S-BAIBA using dual column HPLC-fluorescence. The plasma concentration of baseline R-BAIBA was 67 times higher compared to S-BAIBA (1734 ± 821 vs. 29.3 ± 7.8 nM). Exercise induced a 13 and 20% increase in R-BAIBA and S-BAIBA, respectively. The AGXT2 rs37369 genotype strongly affected baseline levels of R-BAIBA, but did not have an impact on baseline S-BAIBA. We demonstrate that BAIBA should not be treated as one molecule, given (1) the markedly uneven distribution of its enantiomers in human plasma favoring R-BAIBA, and (2) their different metabolic source, as evidenced by the AGXT2 polymorphism only affecting R-BAIBA. The proposed function in organ cross talk is supported by the current data and may apply to both enantiomers, but the tissue of origin remains unclear.
Collapse
Affiliation(s)
- Jan Stautemas
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - André B P Van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Lida Stroomer
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Fred Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Laura Blancquaert
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Filip B D Lefevere
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Inge Everaert
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sports Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Ottosson F, Ericson U, Almgren P, Smith E, Brunkwall L, Hellstrand S, Nilsson PM, Orho-Melander M, Fernandez C, Melander O. Dimethylguanidino Valerate: A Lifestyle-Related Metabolite Associated With Future Coronary Artery Disease and Cardiovascular Mortality. J Am Heart Assoc 2019; 8:e012846. [PMID: 31533499 PMCID: PMC6806048 DOI: 10.1161/jaha.119.012846] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Identification of lifestyle modifiable metabolic pathways related to cardiometabolic disease risk is essential for improvement of primary prevention in susceptible individuals. It was recently shown that plasma dimethylguanidino valerate (DMGV) levels are associated with incident type 2 diabetes mellitus. Our aims were to investigate whether plasma DMGV is related to risk of future coronary artery disease and with cardiovascular mortality and to replicate the association with type 2 diabetes mellitus and pinpoint candidate lifestyle interventions susceptible to modulate DMGV levels. Methods and Results Plasma DMGV levels were measured using liquid chromatography‐mass spectrometry in a total of 5768 participants from the MDC (Malmö Diet and Cancer Study—Cardiovascular Cohort), MPP (Malmö Preventive Project), and MOS (Malmö Offspring Study). Dietary intake assessment was performed in the MOS. Baseline levels of DMGV associated with incident coronary artery disease in both the MDC (hazard ratio=1.29; CI=1.16–1.43; P<0.001) and MPP (odds ratio=1.25; CI=1.08–1.44; P=2.4e‐3). In the MDC, DMGV was associated with cardiovascular mortality and incident coronary artery disease, independently of traditional risk factors. Furthermore, the association between DMGV and incident type 2 diabetes mellitus was replicated in both the MDC (hazard ratio=1.83; CI=1.63–2.05; P<0.001) and MPP (odds ratio=1.65; CI=1.38–1.98; P<0.001). Intake of sugar‐sweetened beverages was associated with increased levels of DMGV, whereas intake of vegetables and level of physical activity was associated with lower DMGV. Conclusions We discovered novel independent associations between plasma DMGV and incident coronary artery disease and cardiovascular mortality, while replicating the previously reported association with incident type 2 diabetes mellitus. Additionally, strong associations with sugar‐sweetened beverages, vegetable intake, and physical activity suggest the potential to modify DMGV levels using lifestyle interventions.
Collapse
Affiliation(s)
- Filip Ottosson
- Department of Clinical Sciences Lund University Malmö Sweden
| | - Ulrika Ericson
- Department of Clinical Sciences Lund University Malmö Sweden
| | - Peter Almgren
- Department of Clinical Sciences Lund University Malmö Sweden
| | - Einar Smith
- Department of Clinical Sciences Lund University Malmö Sweden
| | | | | | - Peter M Nilsson
- Department of Clinical Sciences Lund University Malmö Sweden
| | | | | | - Olle Melander
- Department of Clinical Sciences Lund University Malmö Sweden
| |
Collapse
|
13
|
The Second Life of Methylarginines as Cardiovascular Targets. Int J Mol Sci 2019; 20:ijms20184592. [PMID: 31533264 PMCID: PMC6769906 DOI: 10.3390/ijms20184592] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/13/2019] [Accepted: 09/15/2019] [Indexed: 02/07/2023] Open
Abstract
Endogenous methylarginines were proposed as cardiovascular risk factors more than two decades ago, however, so far, this knowledge has not led to the development of novel therapeutic approaches. The initial studies were primarily focused on the endogenous inhibitors of nitric oxide synthases asymmetric dimethylarginine (ADMA) and monomethylarginine (MMA) and the main enzyme regulating their clearance dimethylarginine dimethylaminohydrolase 1 (DDAH1). To date, all the screens for DDAH1 activators performed with the purified recombinant DDAH1 enzyme have not yielded any promising hits, which is probably the main reason why interest towards this research field has started to fade. The relative contribution of the second DDAH isoenzyme DDAH2 towards ADMA and MMA clearance is still a matter of controversy. ADMA, MMA and symmetric dimethylarginine (SDMA) are also metabolized by alanine: glyoxylate aminotransferase 2 (AGXT2), however, in addition to methylarginines, this enzyme also has several cardiovascular protective substrates, so the net effect of possible therapeutic targeting of AGXT2 is currently unclear. Recent studies on regulation and functions of the enzymes metabolizing methylarginines have given a second life to this research direction. Our review discusses the latest discoveries and controversies in the field and proposes novel directions for targeting methylarginines in clinical settings.
Collapse
|
14
|
Establishment of reference values for the lysine acetylation marker Nɛ-acetyllysine in small volume human plasma samples by a multi-target LC–MS/MS method. Amino Acids 2019; 51:1259-1271. [DOI: 10.1007/s00726-019-02765-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/23/2019] [Indexed: 10/26/2022]
|
15
|
Genetic regulation of dimethylarginines and endothelial dysfunction in rheumatoid arthritis. Amino Acids 2019; 51:983-990. [PMID: 31062169 DOI: 10.1007/s00726-019-02740-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Rheumatoid Arthritis (RA) confers an increased cardiovascular disease (CVD) risk which accounts for much of the premature morbidity and mortality observed in this population. Alterations in vascular function and morphology leading to increased atherosclerotic burden are considered the main drivers of CVD in RA individuals with systemic inflammation playing a key role in the dysregulation of endothelial homeostasis and initiation of vascular injury. Dimethylarginines are endogenous inhibitors of nitric oxide (NO) synthase and have emerged as novel, independent biomarkers of CVD in a wide range of conditions associated with vascular pathology. In RA several reports have demonstrated abnormal dimethylarginine metabolism attributable to various factors such as systemic inflammation, decreased degradation or upregulated synthesis. Although a causal relationship between dimethylarginines and vascular damage in RA has not been established, the tight interrelations between inflammation, dimethylarginines and endothelial dysfunction suggest that determination of dimethylarginine regulators may shed more light in the pathophysiology of the atherosclerotic process in RA and may also provide new therapeutic targets. The Alanine-Glyoxylate Aminotransferase 2 (AGTX2)-dependent pathway is a relatively recently discovered alternative pathway of dimethylarginine catabolism and its role on RA-related atherosclerotic disease is yet to be established. As factors affecting dimethylarginine concentrations linked to CVD risk and endothelial dysfunction are of prominent clinical relevance in RA, we present preliminary evidence that gene variants of AGTX-2 may influence dimethylarginine levels in RA patients and provide the rationale for larger studies in this field.
Collapse
|
16
|
Zhao WC, Li G, Huang CY, Jiang JL. Asymmetric dimethylarginine: An crucial regulator in tissue fibrosis. Eur J Pharmacol 2019; 854:54-61. [PMID: 30951718 DOI: 10.1016/j.ejphar.2019.03.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Fibrosis is a reparative process with very few therapeutic options to prevent its progression to organ dysfunction. Chronic fibrotic diseases contribute to an estimated 45% of all death in the industrialized world. Asymmetric dimethylarginine (ADMA), an endothelial nitric oxide synthase inhibitor, plays a crucial role in the pathogenesis of various cardiovascular diseases associated with endothelial dysfunction. Recent reports have focused on ADMA in the pathogenesis of tissue fibrosis. This review discusses the current knowledge about ADMA biology, its association with risk factors of established fibrotic diseases and the potential pathophysiological mechanisms implicating ADMA in the process of tissue fibrosis.
Collapse
Affiliation(s)
- Wei-Chen Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Ge Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Faculty of Medical Public Courses, Xinhua College of Sun Yat-sen University, Guangzhou, Guangdong, 510520, China
| | - Chu-Yi Huang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Jun-Lin Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, China.
| |
Collapse
|
17
|
Tanianskii DA, Jarzebska N, Birkenfeld AL, O'Sullivan JF, Rodionov RN. Beta-Aminoisobutyric Acid as a Novel Regulator of Carbohydrate and Lipid Metabolism. Nutrients 2019; 11:E524. [PMID: 30823446 PMCID: PMC6470580 DOI: 10.3390/nu11030524] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 01/04/2023] Open
Abstract
The prevalence and incidence of metabolic syndrome is reaching pandemic proportions worldwide, thus warranting an intensive search for novel preventive and treatment strategies. Recent studies have identified a number of soluble factors secreted by adipocytes and myocytes (adipo-/myokines), which link sedentary life style, abdominal obesity, and impairments in carbohydrate and lipid metabolism. In this review, we discuss the metabolic roles of the recently discovered myokine β-aminoisobutyric acid (BAIBA), which is produced by skeletal muscle during physical activity. In addition to physical activity, the circulating levels of BAIBA are controlled by the mitochondrial enzyme alanine: glyoxylate aminotransferase 2 (AGXT2), which is primarily expressed in the liver and kidneys. Recent studies have shown that BAIBA can protect from diet-induced obesity in animal models. It induces transition of white adipose tissue to a "beige" phenotype, which induces fatty acids oxidation and increases insulin sensitivity. While the exact mechanisms of BAIBA-induced metabolic effects are still not well understood, we discuss some of the proposed pathways. The reviewed data provide new insights into the connection between physical activity and energy metabolism and suggest that BAIBA might be a potential novel drug for treatment of the metabolic syndrome and its cardiovascular complications.
Collapse
Affiliation(s)
- Dmitrii A Tanianskii
- Department of Biochemistry, Institute of Experimental Medicine, Acad. Pavlov St., 12, 197376 St. Petersburg, Russia.
- Department of Fundamental Medicine and Medical Technology, St.Petersburg State University, 8 liter A, 21st Line V.O., 199034 St. Petersburg, Russia.
| | - Natalia Jarzebska
- University Center for Vascular Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - Andreas L Birkenfeld
- Medical Clinic III, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| | - John F O'Sullivan
- Medical Clinic III, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
- Charles Perkins Centre and Heart Research Institute, The University of Sydney, 7 Eliza St, Newtown NSW, Sydney 2042, Australia.
| | - Roman N Rodionov
- University Center for Vascular Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
18
|
Hu XL, Li MP, Song PY, Tang J, Chen XP. AGXT2: An unnegligible aminotransferase in cardiovascular and urinary systems. J Mol Cell Cardiol 2017; 113:33-38. [PMID: 28970090 DOI: 10.1016/j.yjmcc.2017.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/22/2017] [Accepted: 09/27/2017] [Indexed: 01/07/2023]
Abstract
Cardiovascular diseases (CVDs) and renal impairment interact in a complex and interdependent manner, which makes clarification of possible pathogenesis between CVDs and renal diseases very challenging and important. There is increasing evidence showing that both asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) play a crucial role in the development of CVDs as well as in the prediction of cardiovascular events. Also, the plasma levels of ADMA and SDMA were reported to be significantly associated with renal function. Alanine-glyoxylate aminotransferase 2 (AGXT2) is reported to be involved in ADMA and SDMA metabolism, thus deficiency in the expression or activity of AGXT2 may play a part in the progression of cardiovascular or renal diseases through affecting ADMA/SDMA levels. Here, we focused our attention on AGXT2 and discussed its potential impact on CVDs and renal diseases. Meanwhile, the review also summarized the functions and recent advances of AGXT2, as well as the clinical association studies of AGXT2 in cardiovascular and urinary systems, which might arouse the interest of researchers in these fields.
Collapse
Affiliation(s)
- Xiao-Lei Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Mu-Peng Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Pei-Yuan Song
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, China.
| |
Collapse
|
19
|
Hu XL, Zeng WJ, Li MP, Yang YL, Kuang DB, Li H, Zhang YJ, Jiang C, Peng LM, Qi H, Zhang K, Chen XP. AGXT2 rs37369 polymorphism predicts the renal function in patients with chronic heart failure. Gene 2017; 637:145-151. [PMID: 28942034 DOI: 10.1016/j.gene.2017.09.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/05/2017] [Accepted: 09/19/2017] [Indexed: 01/21/2023]
Abstract
Patients with chronic heart failure (CHF) are often accompanied with varying degrees of renal diseases. The purpose of this study was to identify rs37369 polymorphism of AGXT2 specific to the renal function of CHF patients. A total of 1012 southern Chinese participants, including 487 CHF patients without history of renal diseases and 525 healthy volunteers, were recruited for this study. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) was used to determine the genotypes of AGXT2 rs37369 polymorphism. Levels of blood urea nitrogen (BUN) and serum creatinine (SCr) were detected to indicate the renal function of the participants. BUN level was significantly higher in CHF patients without history of renal diseases compared with healthy volunteers (p=0.000). And the similar result was also obtained for SCr (p=0.000). Besides, our results indicated that the level of BUN correlated significantly with SCr in both the CHF patients without renal diseases (r=0.4533, p<0.0001) and volunteers (r=0.2489, p<0.0001). Furthermore, we found that the AGXT2 rs37369 polymorphism could significantly affect the level of BUN in CHF patients without history of renal diseases (p=0.036, AA+AG vs GG). Patients with rs37369 GG genotype showed a significantly reduced level of BUN compared to those with the AA genotype (p=0.024), and the significant difference was still observed in the smokers of CHF patients without renal diseases (p=0.023). In conclusion, we found that CHF might induce the impairment of kidney and cause deterioration of renal function. AGXT2 rs37369 polymorphism might affect the renal function of CHF patients free from renal diseases, especially in patients with cigarette smoking.
Collapse
Affiliation(s)
- Xiao-Lei Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wen-Jing Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Mu-Peng Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yong-Long Yang
- Haikou People's Hospital, Affiliated Haikou Hospital of Xiangya Medical School, Central South University, Haikou 570311, China
| | - Da-Bin Kuang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - He Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan-Jiao Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chun Jiang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Li-Ming Peng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hong Qi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ke Zhang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha 410013, China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
20
|
Dimitroulas T, Hodson J, Panoulas VF, Sandoo A, Smith J, Kitas G. Genetic variations in the alanine-glyoxylate aminotransferase 2 (AGXT2) gene and dimethylarginines levels in rheumatoid arthritis. Amino Acids 2017; 49:1133-1141. [PMID: 28357606 DOI: 10.1007/s00726-017-2413-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/22/2017] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is associated with high rates of cardiovascular events mainly due to coronary and cerebrovascular atherosclerotic disease. Asymmetric (ADMA) and symmetric (SDMA) dimethylarginines are endogenous inhibitors of nitric oxide synthase and have been repeatedly linked with adverse cardiovascular outcomes in the general population and various disease settings. Alanine-glyoxylate aminotransferase 2 (AGTX2) is considered an alternative metabolic pathway contributing to the clearance of dimethylarginines in humans. The aim of the current study was to investigate the effect of specific AGXT-2 gene polymorphisms on circulating levels of ADMA or SDMA in patients with RA. Serum ADMA and SDMA levels were measured in 201 individuals with RA [median age: 67 years (IQR: 59-73), 155 females]. Two single nucleotide polymorphisms (SNPs) in the AGXT-2 gene-rs37369 and rs28305-were genotyped. Distributions of SDMA and ADMA were skewed, hence comparisons across the gene polymorphisms were performed using Kruskal-Wallis tests, and summarized using medians and interquartile ranges. Univariable analysis did not demonstrate a significant difference in the levels of SDMA or ADMA amongst the different genotypic groups of either rs37369AGXT2 (p = 0.800, 0.977) or rs28305AGXT2 (p = 0.463, 0.634). In multivariable analyses, ADMA levels were found to be significantly associated with erythrocyte sedimentation rate and estimated glomerular filtration rate, whilst SDMA levels were significantly associated with estimated glomerular filtration rate and quantitative insulin sensitivity check index. After adjustments for these factors, the relationship between the AGXT2 gene variants and both ADMA and SDMA remained non-significant. Our study in a well-characterized RA population did not show an association between serum concentrations of dimethylarginines and genetic variants of the AGXT2 gene.
Collapse
Affiliation(s)
- Theodoros Dimitroulas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK. .,4th Department of Internal Medicine, School of Medicine, Hippokration Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, 54642, Thessaloniki, Greece.
| | - James Hodson
- Institute of Translational Medicine, Queen Elizabeth Hospital Birmingham, University Hospital Birmingham NHS Foundation Trust, Mindelsohn Way, Birmingham, B15 2WB, UK
| | - Vasileios F Panoulas
- Cardiovascular Sciences, Imperial College London, National Heart and Lung Institute, London, UK
| | - Aamer Sandoo
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK.,School of Sport, Health and Exercise Sciences, Bangor University, George Building, Bangor, Gwynedd, LL57 2PZ, Wales, UK
| | - Jacqueline Smith
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, UK
| | - George Kitas
- Arthritis Research UK Epidemiology Unit, University of Manchester, Oxford Road, Manchester, UK
| |
Collapse
|
21
|
Toxic Dimethylarginines: Asymmetric Dimethylarginine (ADMA) and Symmetric Dimethylarginine (SDMA). Toxins (Basel) 2017; 9:toxins9030092. [PMID: 28272322 PMCID: PMC5371847 DOI: 10.3390/toxins9030092] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/04/2017] [Indexed: 02/07/2023] Open
Abstract
Asymmetric and symmetric dimethylarginine (ADMA and SDMA, respectively) are toxic, non-proteinogenic amino acids formed by post-translational modification and are uremic toxins that inhibit nitric oxide (NO) production and play multifunctional roles in many human diseases. Both ADMA and SDMA have emerged as strong predictors of cardiovascular events and death in a range of illnesses. Major progress has been made in research on ADMA-lowering therapies in animal studies; however, further studies are required to fill the translational gap between animal models and clinical trials in order to treat human diseases related to elevated ADMA/SDMA levels. Here, we review the reported impacts of ADMA and SDMA on human health and disease, focusing on the synthesis and metabolism of ADMA and SDMA; the pathophysiological roles of these dimethylarginines; clinical conditions and animal models associated with elevated ADMA and SDMA levels; and potential therapies against ADMA and SDMA. There is currently no specific pharmacological therapy for lowering the levels and counteracting the deleterious effects of ADMA and SDMA. A better understanding of the mechanisms underlying the impact of ADMA and SDMA on a wide range of human diseases is essential to the development of specific therapies against diseases related to ADMA and SDMA.
Collapse
|
22
|
Asymmetric Dimethylarginine and Hepatic Encephalopathy: Cause, Effect or Association? Neurochem Res 2016; 42:750-761. [PMID: 27885576 PMCID: PMC5357500 DOI: 10.1007/s11064-016-2111-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/07/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022]
Abstract
The methylated derivative of l-arginine, asymmetric dimethylarginine (ADMA) is synthesized in different mammalian tissues including the brain. ADMA acts as an endogenous, nonselective, competitive inhibitor of all three isoforms of nitric oxide synthase (NOS) and may limit l-arginine supply from the plasma to the enzyme via reducing its transport by cationic amino acid transporters. Hepatic encephalopathy (HE) is a relatively frequently diagnosed complex neuropsychiatric syndrome associated with acute or chronic liver failure, characterized by symptoms linked with impaired brain function leading to neurological disabilities. The l-arginine—nitric oxide (NO) pathway is crucially involved in the pathomechanism of HE via modulating important cerebral processes that are thought to contribute to the major HE symptoms. Specifically, activation of this pathway in acute HE leads to an increase in NO production and free radical formation, thus, contributing to astrocytic swelling and cerebral edema. Moreover, the NO-cGMP pathway seems to be involved in cerebral blood flow (CBF) regulation, altered in HE. For this reason, depressed NO-cGMP signaling accompanying chronic HE and ensuing cGMP deficit contributes to the cognitive and motor failure. However, it should be remembered that ADMA, a relatively little known element limiting NO synthesis in HE, may also influence the NO-cGMP pathway regulation. In this review, we will discuss the contribution of ADMA to the regulation of the NO-cGMP pathway in the brain, correlation of ADMA level with CBF and cognitive alterations observed during HE progression in patients and/or animal models of HE.
Collapse
|
23
|
Vemula H, Kitase Y, Ayon NJ, Bonewald L, Gutheil WG. Gaussian and linear deconvolution of LC-MS/MS chromatograms of the eight aminobutyric acid isomers. Anal Biochem 2016; 516:75-85. [PMID: 27771391 DOI: 10.1016/j.ab.2016.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
Isomeric molecules present a challenge for analytical resolution and quantification, even with MS-based detection. The eight aminobutyric acid (ABA) isomers are of interest for their various biological activities, particularly γ-aminobutyric acid (GABA) and the d- and l-isomers of β-aminoisobutyric acid (β-AIBA; BAIBA). This study aimed to investigate LC-MS/MS-based resolution of these ABA isomers as their Marfey's (Mar) reagent derivatives. HPLC was able to separate three Mar-ABA isomers l-β-ABA (l-BABA), and l- and d-α-ABA (AABA) completely, with three isomers (GABA, and d/l-BAIBA) in one chromatographic cluster, and two isomers (α-AIBA (AAIBA) and d-BABA) in a second cluster. Partially separated cluster components were deconvoluted using Gaussian peak fitting except for GABA and d-BAIBA. MS/MS detection of Marfey's derivatized ABA isomers provided six MS/MS fragments, with substantially different intensity profiles between structural isomers. This allowed linear deconvolution of ABA isomer peaks. Combining HPLC separation with linear and Gaussian deconvolution allowed resolution of all eight ABA isomers. Application to human serum found a substantial level of l-AABA (13 μM), an intermediate level of l-BAIBA (0.8 μM), and low but detectable levels (<0.2 μM) of GABA, l-BABA, AAIBA, d-BAIBA, and d-AABA. This approach should be useful for LC-MS/MS deconvolution of other challenging groups of isomeric molecules.
Collapse
Affiliation(s)
- Harika Vemula
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yukiko Kitase
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Navid J Ayon
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Lynda Bonewald
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - William G Gutheil
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
24
|
Burdin DV, Kolobov AA, Brocker C, Soshnev AA, Samusik N, Demyanov AV, Brilloff S, Jarzebska N, Martens-Lobenhoffer J, Mieth M, Maas R, Bornstein SR, Bode-Böger SM, Gonzalez F, Weiss N, Rodionov RN. Diabetes-linked transcription factor HNF4α regulates metabolism of endogenous methylarginines and β-aminoisobutyric acid by controlling expression of alanine-glyoxylate aminotransferase 2. Sci Rep 2016; 6:35503. [PMID: 27752141 PMCID: PMC5067591 DOI: 10.1038/srep35503] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/30/2016] [Indexed: 02/07/2023] Open
Abstract
Elevated levels of circulating asymmetric and symmetric dimethylarginines (ADMA and SDMA) predict and potentially contribute to end organ damage in cardiovascular diseases. Alanine-glyoxylate aminotransferase 2 (AGXT2) regulates systemic levels of ADMA and SDMA, and also of beta-aminoisobutyric acid (BAIB)-a modulator of lipid metabolism. We identified a putative binding site for hepatic nuclear factor 4 α (HNF4α) in AGXT2 promoter sequence. In a luciferase reporter assay we found a 75% decrease in activity of Agxt2 core promoter after disruption of the HNF4α binding site. Direct binding of HNF4α to Agxt2 promoter was confirmed by chromatin immunoprecipitation assay. siRNA-mediated knockdown of Hnf4a led to an almost 50% reduction in Agxt2 mRNA levels in Hepa 1–6 cells. Liver-specific Hnf4a knockout mice exhibited a 90% decrease in liver Agxt2 expression and activity, and elevated plasma levels of ADMA, SDMA and BAIB, compared to wild-type littermates. Thus we identified HNF4α as a major regulator of Agxt2 expression. Considering a strong association between human HNF4A polymorphisms and increased risk of type 2 diabetes our current findings suggest that downregulation of AGXT2 and subsequent impairment in metabolism of dimethylarginines and BAIB caused by HNF4α deficiency might contribute to development of cardiovascular complications in diabetic patients.
Collapse
Affiliation(s)
- Dmitry V Burdin
- Department of Physiology, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| | - Alexey A Kolobov
- Department of Biochemistry, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| | - Chad Brocker
- National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | | | - Nikolay Samusik
- Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anton V Demyanov
- Institute of Highly Pure Biopreparations, 197110 Saint Petersburg, Russia
| | - Silke Brilloff
- University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Natalia Jarzebska
- University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Maren Mieth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Frank Gonzalez
- National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Norbert Weiss
- University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Roman N Rodionov
- University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
25
|
A Novel Pathway for Metabolism of the Cardiovascular Risk Factor Homoarginine by alanine:glyoxylate aminotransferase 2. Sci Rep 2016; 6:35277. [PMID: 27752063 PMCID: PMC5082758 DOI: 10.1038/srep35277] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/23/2016] [Indexed: 01/06/2023] Open
Abstract
Low plasma concentrations of L-homoarginine are associated with an increased risk of cardiovascular events, while homoarginine supplementation is protective in animal models of metabolic syndrome and stroke. Catabolism of homoarginine is still poorly understood. Based on the recent findings from a Genome Wide Association Study we hypothesized that homoarginine can be metabolized by alanine:glyoxylate aminotransferase 2 (AGXT2). We purified human AGXT2 from tissues of AGXT2 transgenic mice and demonstrated its ability to metabolize homoarginine to 6-guanidino-2-oxocaproic acid (GOCA). After incubation of HepG2 cells overexpressing AGXT2 with isotope-labeled homoarginine-d4 we were able to detect labeled GOCA in the medium. We injected wild type mice with labeled homoarginine and detected labeled GOCA in the plasma. We found that AGXT2 knockout (KO) mice have higher homoarginine and lower GOCA plasma levels as compared to wild type mice, while the reverse was true for AGXT2 transgenic (Tg) mice. In summary, we experimentally proved the presence of a new pathway of homoarginine catabolism – its transamination by AGXT2 with formation of GOCA and demonstrated that endogenous AGXT2 is required for maintenance of homoarginine levels in mice. Our findings may lead to development of novel therapeutic approaches for cardiovascular pathologies associated with homoarginine deficiency.
Collapse
|
26
|
|
27
|
Hu XL, Zhou JP, Kuang DB, Qi H, Peng LM, Yang TL, Li X, Zhang W, Zhou HH, Chen XP. Considerable impacts of AGXT2 V140I polymorphism on chronic heart failure in the Chinese population. Atherosclerosis 2016; 251:255-262. [DOI: 10.1016/j.atherosclerosis.2016.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 06/25/2016] [Accepted: 07/06/2016] [Indexed: 01/02/2023]
|
28
|
Affiliation(s)
- Jessica A. Hokamp
- Department of Veterinary Pathobiology; College of Veterinary Medicine and Biomedical Sciences; Texas A&M University; College Station TX USA
| | - Mary B. Nabity
- Department of Veterinary Pathobiology; College of Veterinary Medicine and Biomedical Sciences; Texas A&M University; College Station TX USA
| |
Collapse
|
29
|
Rodionov RN, Jarzebska N, Weiss N, Lentz SR. AGXT2: a promiscuous aminotransferase. Trends Pharmacol Sci 2014; 35:575-82. [PMID: 25294000 DOI: 10.1016/j.tips.2014.09.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 12/11/2022]
Abstract
Alanine-glyoxylate aminotransferase 2 (AGXT2) is a multifunctional mitochondrial aminotransferase that was first identified in 1978. The physiological importance of AGXT2 was largely overlooked for three decades because AGXT2 is less active in glyoxylate metabolism than AGXT1, the enzyme that is deficient in primary hyperoxaluria type I. Recently, several novel functions of AGXT2 have been 'rediscovered' in the setting of modern genomic and metabolomic studies. It is now apparent that AGXT2 has multiple substrates and products and that altered AGXT2 activity may contribute to the pathogenesis of cardiovascular, renal, neurological, and hematological diseases. This article reviews the biochemical properties and physiological functions of AGXT2, its unique role at the intersection of key mitochondrial pathways, and its potential as a drug target.
Collapse
Affiliation(s)
- Roman N Rodionov
- University Center for Vascular Medicine and Department of Internal Medicine, Division of Angiology, University Hospital 'Carl Gustav Carus', Technische Universität Dresden, Fetscherstrasse 42, 01307 Dresden, Germany
| | - Natalia Jarzebska
- University Hospital 'Carl Gustav Carus', Technische Universität Dresden, Fetscherstrasse 42, 01307 Dresden, Germany
| | - Norbert Weiss
- University Center for Vascular Medicine and Department of Internal Medicine, Division of Angiology, University Hospital 'Carl Gustav Carus', Technische Universität Dresden, Fetscherstrasse 42, 01307 Dresden, Germany
| | - Steven R Lentz
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| |
Collapse
|
30
|
Popolo A, Adesso S, Pinto A, Autore G, Marzocco S. L-Arginine and its metabolites in kidney and cardiovascular disease. Amino Acids 2014; 46:2271-86. [PMID: 25161088 DOI: 10.1007/s00726-014-1825-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 08/13/2014] [Indexed: 12/19/2022]
Abstract
L-Arginine is a semi essential amino acid synthesised from glutamine, glutamate and proline via the intestinal-renal axis in humans and most mammals. L-Arginine degradation occurs via multiple pathways initiated by arginase, nitric-oxide synthase, Arg: glycine amidinotransferase, and Arg decarboxylase. These pathways produce nitric oxide, polyamines, proline, glutamate, creatine and agmatine with each having enormous biological importance. Several disease are associated to an L-arginine impaired levels and/or to its metabolites: in particular various L-arginine metabolites may participate in pathogenesis of kidney and cardiovascular disease. L-Arginine and its metabolites may constitute both a marker of pathology progression both the rationale for manipulating L-arginine metabolism as a strategy to ameliorate these disease. A large number of studies have been performed in experimental models of kidney disease with sometimes conflicting results, which underlie the complexity of Arg metabolism and our incomplete knowledge of all the mechanisms involved. Moreover several lines of evidence demonstrate the role of L-arg metabolites in cardiovascular disease and that L-arg administration role in reversing endothelial dysfunction, which is the leading cause of cardiovascular diseases, such as hypertension and atherosclerosis. This review will discuss the implication of the mains L-arginine metabolites and L-arginine-derived guanidine compounds in kidney and cardiovascular disease considering the more recent literature in the field.
Collapse
Affiliation(s)
- Ada Popolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, SA, Italy
| | | | | | | | | |
Collapse
|
31
|
Rodionov RN, Martens-Lobenhoffer J, Brilloff S, Hohenstein B, Jarzebska N, Jabs N, Kittel A, Maas R, Weiss N, Bode-Böger SM. Role of alanine:glyoxylate aminotransferase 2 in metabolism of asymmetric dimethylarginine in the settings of asymmetric dimethylarginine overload and bilateral nephrectomy. Nephrol Dial Transplant 2014; 29:2035-42. [PMID: 25002409 DOI: 10.1093/ndt/gfu236] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Asymmetric and symmetric dimethylarginines (ADMA and SDMA) predict complications and mortality in cardiovascular and renal diseases. Alanine:glyoxylate aminotransferase 2 (AGXT2) can metabolize both ADMA and SDMA; however, this metabolic pathway is still poorly understood. The goal of our study was to test the hypothesis that AGXT2 is compensatory upregulated in the settings of ADMA overload and bilateral nephrectomy. METHODS ADMA was infused for 3 days using osmotic minipumps in mice. Half of the mice underwent bilateral nephrectomy 24 h before the end of the infusion. RESULTS Infusion of ADMA caused a 3- to 4-fold increase in plasma and urine ADMA levels and a 2- to 3-fold increase in plasma and urine levels of the ADMA-specific metabolite of AGXT2 α-keto-δ-(N,N-dimethylguanidino)valeric acid (DMGV). Bilateral nephrectomy led to an ∼4-fold increase of plasma SDMA levels, but did not change plasma ADMA levels. Interestingly, plasma levels of DMGV were elevated 32-fold in the mice, which underwent bilateral nephrectomy. Neither bilateral nephrectomy nor ADMA infusion caused upregulation of AGXT2 expression or activity. CONCLUSIONS Our data demonstrate that short-term elevation of systemic levels of ADMA leads to a dramatic increase of DMGV formation without upregulation of AGXT2 expression or activity, which suggests that AGXT2-mediated pathway of ADMA metabolism is not saturated under normal conditions and may play a major role in the maintenance of ADMA homeostasis in the setting of local or systemic elevation of ADMA levels.
Collapse
Affiliation(s)
- Roman N Rodionov
- University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Silke Brilloff
- University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bernd Hohenstein
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Natalia Jarzebska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Normund Jabs
- University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anja Kittel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Norbert Weiss
- University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
32
|
Zhou JP, Bai YP, Hu XL, Kuang DB, Shi RZ, Xiong Y, Zhang W, Xia J, Chen BL, Yang TL, Chen XP. Association of the AGXT2 V140I polymorphism with risk for coronary heart disease in a Chinese population. J Atheroscler Thromb 2014; 21:1022-30. [PMID: 24834905 DOI: 10.5551/jat.23077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Asymmetric dimethylarginine (ADMA) is a nitric oxide synthase (NOS) inhibitor that decreases NO production and promotes the development of cardiovascular diseases. Alanine-glyoxylate aminotransferase 2 (AGXT2) plays an important role in ADMA metabolism. This study was designed to explore the association of the AGXT2 V140I (rs37369 G>A) polymorphism with risk for coronary heart disease (CHD) in a Chinese population. METHODS A case-control study including 1103 controls and 942 CHD patients was performed. The patients were genotyped for rs37369 using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Plasma ADMA concentration in healthy controls was measured by an enzyme-linked immunosorbent assay (ELISA). RESULTS The rs37369 GG genotype was significantly overrepresented in CHD patients compared to the controls (18.5% versus 14.8%, p=0.025), and it was significantly associated with increased risk for CHD in smokers (OR=2.21, 95% CI: 1.24-3.92, p=0.007) and marginally increased CHD risk for individuals with diabetes mellitus (OR=1.92; 95% CI: 0.94-3.91, p=0.074). The association between rs37369 and CHD risk was further increased in smokers with diabetes mellitus (OR=3.32, 95% CI:1.14-9.67, p=0.028). Patients who smoked and were rs37369 GG homozygous showed significantly higher plasma ADMA levels than carriers of the rs37369 A allele (p=0.004). However, in non-smokers, patients homozygous for rs37369 GG showed significantly lower plasma ADMA concentrations than carriers of the rs37369 A allele (p=0.003). Furthermore, smokers homozygous for rs37369 GG showed significantly higher plasma ADMA concentrations than non-smokers with the same genotype (p=0.012). CONCLUSION The AGXT2 rs37369 polymorphism is associated with increased risk for CHD in smokers and in diabetes mellitus patients. This increased risk may be due to increased plasma ADMA levels.
Collapse
Affiliation(s)
- Ji-Peng Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kittel A, Müller F, König J, Mieth M, Sticht H, Zolk O, Kralj A, Heinrich MR, Fromm MF, Maas R. Alanine-glyoxylate aminotransferase 2 (AGXT2) polymorphisms have considerable impact on methylarginine and β-aminoisobutyrate metabolism in healthy volunteers. PLoS One 2014; 9:e88544. [PMID: 24586340 PMCID: PMC3933329 DOI: 10.1371/journal.pone.0088544] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 01/06/2014] [Indexed: 12/18/2022] Open
Abstract
Elevated plasma concentrations of asymmetric (ADMA) and symmetric (SDMA) dimethylarginine have repeatedly been linked to adverse clinical outcomes. Both methylarginines are substrates of alanine-glyoxylate aminotransferase 2 (AGXT2). It was the aim of the present study to simultaneously investigate the functional relevance and relative contributions of common AGXT2 single nucleotide polymorphisms (SNPs) to plasma and urinary concentrations of methylarginines as well as β-aminoisobutyrate (BAIB), a prototypic substrate of AGXT2. In a cohort of 400 healthy volunteers ADMA, SDMA and BAIB concentrations were determined in plasma and urine using HPLC-MS/MS and were related to the coding AGXT2 SNPs rs37369 (p.Val140Ile) and rs16899974 (p.Val498Leu). Volunteers heterozygous or homozygous for the AGXT2 SNP rs37369 had higher SDMA plasma concentrations by 5% and 20% (p = 0.002) as well as higher BAIB concentrations by 54% and 146%, respectively, in plasma and 237% and 1661%, respectively, in urine (both p<0.001). ADMA concentrations were not affected by both SNPs. A haplotype analysis revealed that the second investigated AGXT2 SNP rs16899974, which was not significantly linked to the other AGXT2 SNP, further aggravates the effect of rs37369 with respect to BAIB concentrations in plasma and urine. To investigate the impact of the amino acid exchange p.Val140Ile, we established human embryonic kidney cell lines stably overexpressing wild-type or mutant (p.Val140Ile) AGXT2 protein and assessed enzyme activity using BAIB and stable-isotope labeled [²H₆]-SDMA as substrate. In vitro, the amino acid exchange of the mutant protein resulted in a significantly lower enzyme activity compared to wild-type AGXT2 (p<0.05). In silico modeling of the SNPs indicated reduced enzyme stability and substrate binding. In conclusion, SNPs of AGXT2 affect plasma as well as urinary BAIB and SDMA concentrations linking methylarginine metabolism to the common genetic trait of hyper-β-aminoisobutyric aciduria.
Collapse
Affiliation(s)
- Anja Kittel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maren Mieth
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Zolk
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ana Kralj
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - Markus R. Heinrich
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - Martin F. Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Schepers E, Speer T, Bode-Böger SM, Fliser D, Kielstein JT. Dimethylarginines ADMA and SDMA: the real water-soluble small toxins? Semin Nephrol 2014; 34:97-105. [PMID: 24780466 DOI: 10.1016/j.semnephrol.2014.02.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Uremia occurs if the kidney loses the ability to eliminate toxic compounds at a sufficient rate into the urine. In 1970, N-N, N-G- and N-G,N׳-G-dimethyl-arginine (asymmetric dimethylarginine [ADMA] and symmetric dimethylarginine) were isolated from human urine. It was anticipated that both substances might be important in the pathophysiology and for the diagnosis of various pathologic states. It took 22 years, however, before this idea materialized when it was found that ADMA, which is increased in hemodialysis patients, inhibits the synthesis of the endothelial-derived relaxing factor, identified as nitric oxide. ADMA correlates with traditional and nontraditional cardiovascular risk factors and is a strong predictor of cardiovascular events and death in both patients with chronic kidney disease and in the general population. It also seems to mediate adverse cardiovascular effects of drugs such as proton pump inhibitors. To date, we have no specific pharmacologic therapy at hand to neutralize the deleterious effects of ADMA, curbing the enthusiasm for this marker and mediator of cardiovascular disease.
Collapse
Affiliation(s)
| | - Timo Speer
- Department of Internal Medicine IV, Renal and Hypertensive Disease, Saarland University, Medical Centre, Homburg/Saar, Germany
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto von-Guericke University, Magdeburg, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Renal and Hypertensive Disease, Saarland University, Medical Centre, Homburg/Saar, Germany
| | - Jan T Kielstein
- Department of Nephrology and Hypertension, Medical School Hannover, Germany.
| |
Collapse
|
35
|
Martens-Lobenhoffer J, Bode-Böger SM. Mass spectrometric quantification of L-arginine and its pathway related substances in biofluids: the road to maturity. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 964:89-102. [PMID: 24210895 DOI: 10.1016/j.jchromb.2013.10.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/17/2013] [Accepted: 10/19/2013] [Indexed: 11/18/2022]
Abstract
The amino acid L-arginine together with its metabolites and related substances is in the center of many biologically important pathways, especially the urea cycle and the nitric oxide (NO) synthesis. Therefore, the concentrations of these substances in various biological fluids are of great interest as predictive markers for health and disease. Yet, they provide major analytical difficulties as they are very polar in nature and therefore not easily to be separated on standard reversed phase HPLC stationary phases. Furthermore, as endogenous substances, no analyte-free matrix is available, a fact that results in complicated calibration procedures. This review evaluates the analytical literature for the determination of L-arginine, symmetric dimethylarginine, asymmetric dimethylarginine, monomethylarginine, L-citrulline, L-ornithine, L-homoarginine, agmatine and dimethylguanidinovaleric acid in biological fluids. Papers are discussed, which were published since 2007 and describe methods applying capillary electrophoresis (CE), gas chromatography (GC), reversed phase HPLC or polar phase HPLC, coupled to mass spectrometric quantification. Nowadays, many carefully developed and validated methods for L-arginine and its related substances are available to the scientific community. The use of stable isotope labeled internal standards enables high precision and accuracy in mass spectrometry-based quantitative analysis.
Collapse
Affiliation(s)
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
36
|
Seppälä I, Kleber ME, Lyytikäinen LP, Hernesniemi JA, Mäkelä KM, Oksala N, Laaksonen R, Pilz S, Tomaschitz A, Silbernagel G, Boehm BO, Grammer TB, Koskinen T, Juonala M, Hutri-Kähönen N, Alfthan G, Viikari JSA, Kähonen M, Raitakari OT, März W, Meinitzer A, Lehtimäki T. Genome-wide association study on dimethylarginines reveals novel AGXT2 variants associated with heart rate variability but not with overall mortality. Eur Heart J 2013; 35:524-31. [PMID: 24159190 DOI: 10.1093/eurheartj/eht447] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS The purpose of this study was to identify novel genetic variants influencing circulating asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA) levels and to evaluate whether they have a prognostic value on cardiovascular mortality. METHODS AND RESULTS We conducted a genome-wide association study on the methylarginine traits and investigated the predictive value of the new discovered variants on mortality. Our meta-analyses replicated the previously known locus for ADMA levels in DDAH1 (rs997251; P = 1.4 × 10(-40)), identified two non-synomyous polymorphisms for SDMA levels in AGXT2 (rs37369; P = 1.4 × 10(-40) and rs16899974; P = 1.5 × 10(-38)) and one in SLC25A45 (rs34400381; P = 2.5 × 10(-10)). We also fine-mapped the AGXT2 locus for further independent association signals. The two non-synonymous AGXT2 variants independently associated with SDMA levels were also significantly related with short-term heart rate variability (HRV) indices in young adults. The major allele (C) of the novel non-synonymous rs16899974 (V498L) variant associated with decreased SDMA levels and an increase in the ratio between the low- and high-frequency spectral components of HRV (P = 0.00047). Furthermore, the SDMA decreasing allele (G) of the non-synomyous SLC25A45 (R285C) variant was associated with a lower resting mean heart rate during the HRV measurements (P = 0.0046), but not with the HRV indices. None of the studied genome-wide significant variants had any major effect on cardiovascular or total mortality in patients referred for coronary angiography. CONCLUSIONS AGXT2 has an important role in SDMA metabolism in humans. AGXT2 may additionally have an unanticipated role in the autonomic nervous system regulation of cardiac function.
Collapse
Affiliation(s)
- Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University School of Medicine, Finn-Medi 2, 3rd floor, PO Box 2000, Tampere FI-33521, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|