1
|
Jarzebska N, Bornstein SR, Tselmin S, Julius U, Cellini B, Siow R, Martin M, Mookerjee RP, Mangoni AA, Weiss N, Rodionov R. Asymmetric Dimethylarginine: A Never-Aging Story. Horm Metab Res 2025. [PMID: 40418971 DOI: 10.1055/a-2537-4692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Human aging is intrinsically associated with the onset and the progression of several disease states causing significant disability and poor quality of life. Although such association was traditionally considered immutable, recent advances have led to a better understanding of several critical biochemical pathways involved in the aging process. This, in turn, has stimulated a significant body of research to investigate whether reprogramming these pathways could delay the progression of human ageing and/or prevent relevant disease states, ultimately favoring healthier aging process. Cellular senescence is regarded as the principal causative factor implicated in biological and pathophysiological processes involved in aging. Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthase and an independent risk factor for several age-associated diseases. The selective extracorporeal removal of ADMA is emerging as a promising strategy to reduce the burden of age-associated disease states. This article discusses the current knowledge regarding the critical pathways involved in human aging and associated diseases and the possible role of ADMA as a target for therapies leading to healthier aging processes.
Collapse
Affiliation(s)
- Natalia Jarzebska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Sergey Tselmin
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Ulrich Julius
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Richard Siow
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Ageing Research at King's (ARK), King's College London, London, United Kingdom of Great Britain and Northern Ireland
- Department of Physiology, Anatomy and Genetics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Mike Martin
- Department of Psychology, University of Zurich, Zurich, Switzerland
- Healthy Longevity Center, University of Zurich, Zurich, Switzerland
| | - Rajeshwar P Mookerjee
- Institute of Liver and Digestive Health, University College London, London, United Kingdom of Great Britain and Northern Ireland
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| | - Norbert Weiss
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Roman Rodionov
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, Australia
| |
Collapse
|
2
|
Prosekina EA, Shapkina VA, Karpov AE, Fedorutseva EY, Artemyeva AS. [DDAH1 protein: biological functions, role in carcinogenesis processes]. Arkh Patol 2025; 87:60-67. [PMID: 39943731 DOI: 10.17116/patol20258701160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Dimethylarginine Dimethylaminohydrolase 1 (DDAH1) is an essential enzyme capable of degrading asymmetric dimethylarginine, which is an endogenous inhibitor of nitric oxide synthase. Increased expression of DDAH1 and subsequent increased NO production are associated with carcinogenesis. In particular, DDAH1 is involved in the creation of a vascular network by tumor cells, vasculogenic mimicry, which is closely associated with tumor progression and poor patient prognosis. This is the reason why DDAH1 may be a potential therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- E A Prosekina
- N.N. Petrov National Medical Research Center of Oncology, St. Petersburg, Russia
- National Research Tomsk State University, Tomsk, Russia
| | - V A Shapkina
- St. Petersburg State University, St. Petersburg, Russia
| | - A E Karpov
- N.N. Petrov National Medical Research Center of Oncology, St. Petersburg, Russia
| | | | - A S Artemyeva
- N.N. Petrov National Medical Research Center of Oncology, St. Petersburg, Russia
| |
Collapse
|
3
|
Nair PC, Mangoni AA, Rodionov RN. Redefining the biological and pathophysiological role of dimethylarginine dimethylaminohydrolase 2. Trends Mol Med 2024; 30:552-561. [PMID: 38553332 DOI: 10.1016/j.molmed.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 06/15/2024]
Abstract
The enzyme dimethylarginine dimethylaminohydrolase (DDAH) 1 metabolizes asymmetric dimethylarginine (ADMA), a critical endogenous cardiovascular risk factor. In the past two decades, there has been significant controversy about whether DDAH2, the other DDAH isoform, is also able to directly metabolize ADMA. There has been evidence that DDAH2 regulates several critical processes involved in cardiovascular and immune homeostasis. However, the molecular mechanisms underpinning these effects are unclear. In this opinion, we discuss the previous and current knowledge of ADMA metabolism by DDAH in light of a recent consortium study, which convincingly demonstrated that DDAH2 is not capable of metabolizing ADMA, unlike DDAH1. Thus, further research in this field is needed to uncover the molecular mechanisms of DDAH2 and its role in various disorders.
Collapse
Affiliation(s)
- Pramod C Nair
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia; Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia; South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, SA, Australia; Discipline of Medicine, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA, Australia
| | - Roman N Rodionov
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
4
|
Kozlova AA, Rubets E, Vareltzoglou MR, Jarzebska N, Ragavan VN, Chen Y, Martens-Lobenhoffer J, Bode-Böger SM, Gainetdinov RR, Rodionov RN, Bernhardt N. Knock-out of the critical nitric oxide synthase regulator DDAH1 in mice impacts amphetamine sensitivity and dopamine metabolism. J Neural Transm (Vienna) 2023; 130:1097-1112. [PMID: 36792833 PMCID: PMC10460711 DOI: 10.1007/s00702-023-02597-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/28/2023] [Indexed: 02/17/2023]
Abstract
The enzyme dimethylarginine dimethylaminohydrolase 1 (DDAH1) plays a pivotal role in the regulation of nitric oxide levels by degrading the main endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA). Growing evidence highlight the potential implication of DDAH/ADMA axis in the etiopathogenesis of several neuropsychiatric and neurological disorders, yet the underlying molecular mechanisms remain elusive. In this study, we sought to investigate the role of DDAH1 in behavioral endophenotypes with neuropsychiatric relevance. To achieve this, a global DDAH1 knock-out (DDAH1-ko) mouse strain was employed. Behavioral testing and brain region-specific neurotransmitter profiling have been conducted to assess the effect of both genotype and sex. DDAH1-ko mice exhibited increased exploratory behavior toward novel objects, altered amphetamine response kinetics and decreased dopamine metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) level in the piriform cortex and striatum. Females of both genotypes showed the most robust amphetamine response. These results support the potential implication of the DDAH/ADMA pathway in central nervous system processes shaping the behavioral outcome. Yet, further experiments are required to complement the picture and define the specific brain-regions and mechanisms involved.
Collapse
Affiliation(s)
- Alena A Kozlova
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Elena Rubets
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Magdalini R Vareltzoglou
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Vinitha N Ragavan
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Yingjie Chen
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | | | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-Von-Guericke University, Magdeburg, Germany
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine and Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034, Saint-Petersburg, Russia
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307, Dresden, Germany
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany.
| |
Collapse
|
5
|
Ragavan VN, Nair PC, Jarzebska N, Angom RS, Ruta L, Bianconi E, Grottelli S, Tararova ND, Ryazanskiy D, Lentz SR, Tommasi S, Martens-Lobenhoffer J, Suzuki-Yamamoto T, Kimoto M, Rubets E, Chau S, Chen Y, Hu X, Bernhardt N, Spieth PM, Weiss N, Bornstein SR, Mukhopadhyay D, Bode-Böger SM, Maas R, Wang Y, Macchiarulo A, Mangoni AA, Cellini B, Rodionov RN. A multicentric consortium study demonstrates that dimethylarginine dimethylaminohydrolase 2 is not a dimethylarginine dimethylaminohydrolase. Nat Commun 2023; 14:3392. [PMID: 37296100 PMCID: PMC10256801 DOI: 10.1038/s41467-023-38467-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/27/2023] [Indexed: 06/12/2023] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects against cardiovascular disease by metabolising the risk factor asymmetric dimethylarginine (ADMA). However, the question whether the second DDAH isoform, DDAH2, directly metabolises ADMA has remained unanswered. Consequently, it is still unclear if DDAH2 may be a potential target for ADMA-lowering therapies or if drug development efforts should focus on DDAH2's known physiological functions in mitochondrial fission, angiogenesis, vascular remodelling, insulin secretion, and immune responses. Here, an international consortium of research groups set out to address this question using in silico, in vitro, cell culture, and murine models. The findings uniformly demonstrate that DDAH2 is incapable of metabolising ADMA, thus resolving a 20-year controversy and providing a starting point for the investigation of alternative, ADMA-independent functions of DDAH2.
Collapse
Affiliation(s)
- Vinitha N Ragavan
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Pramod C Nair
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
- Flinders Health and Medical Research Institute (FHMRI), College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Cancer Program, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, SA, Australia
- Discipline of Medicine, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Natalia Jarzebska
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Luana Ruta
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Silvia Grottelli
- Department of Medicine and Surgery, University of Perugia, P.le L. Sevari 1, Perugia, Italy
| | | | | | - Steven R Lentz
- Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sara Tommasi
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | | | - Toshiko Suzuki-Yamamoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan
| | - Masumi Kimoto
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Okayama, Japan
| | - Elena Rubets
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Sarah Chau
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, NY, USA
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xinli Hu
- Institute of Molecular Medicine, Beijing University, Beijing, China
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Peter M Spieth
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Norbert Weiss
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
- School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto von Guericke University, Magdeburg, Germany
| | - Renke Maas
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU New - Research Center for New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, NY, USA
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, via del Liceo 1, Perugia, Italy
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, Adelaide, SA, Australia
| | - Barbara Cellini
- Department of Medicine and Surgery, University of Perugia, P.le L. Sevari 1, Perugia, Italy
| | - Roman N Rodionov
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany.
- College of Medicine and Public Health, Flinders University and Flinders Medical Center, Adelaide, SA, Australia.
| |
Collapse
|
6
|
Parthasarathy S, Soundararajan P, Sakthivelu M, Karuppiah KM, Velusamy P, Gopinath SC, Pachaiappan R. The role of prognostic biomarkers and their implications in early detection of preeclampsia: A systematic review. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
7
|
Chen X, Xu J, Bao W, Li H, Wu W, Liu J, Pi J, Tomlinson B, Chan P, Ruan C, Zhang Q, Zhang L, Fan H, Morrisey E, Liu Z, Zhang Y, Lin L, Liu J, Zhuang T. Endothelial Foxp1 Regulates Neointimal Hyperplasia Via Matrix Metalloproteinase-9/Cyclin Dependent Kinase Inhibitor 1B Signal Pathway. J Am Heart Assoc 2022; 11:e026378. [PMID: 35904197 PMCID: PMC9375493 DOI: 10.1161/jaha.122.026378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The endothelium is essential for maintaining vascular physiological homeostasis and the endothelial injury leads to the neointimal hyperplasia because of the excessive proliferation of vascular smooth muscle cells. Endothelial Foxp1 (forkhead box P1) has been shown to control endothelial cell (EC) proliferation and migration in vitro. However, whether EC-Foxp1 participates in neointimal formation in vivo is not clear. Our study aimed to investigate the roles and mechanisms of EC-Foxp1 in neointimal hyperplasia. Methods and Results The wire injury femoral artery neointimal hyperplasia model was performed in Foxp1 EC-specific loss-of-function and gain-of-function mice. EC-Foxp1 deletion mice displayed the increased neointimal formation through elevation of vascular smooth muscle cell proliferation and migration, and the reduction of EC proliferation hence reendothelialization after injury. In contrast, EC-Foxp1 overexpression inhibited the neointimal formation. EC-Foxp1 paracrine regulated vascular smooth muscle cell proliferation and migration via targeting matrix metalloproteinase-9. Also, EC-Foxp1 deletion impaired EC repair through reduction of EC proliferation via increasing cyclin dependent kinase inhibitor 1B expression. Delivery of cyclin dependent kinase inhibitor 1B-siRNA to ECs using RGD (Arg-Gly-Asp)-peptide magnetic nanoparticle normalized the EC-Foxp1 deletion-mediated impaired EC repair and attenuated the neointimal formation. EC-Foxp1 regulates matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B signaling pathway to control injury induced neointimal formation. Conclusions Our study reveals that targeting EC-Foxp1-matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B pathway might provide future novel therapeutic interventions for restenosis.
Collapse
Affiliation(s)
- Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jianfei Xu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenzhen Bao
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Hongda Li
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenrun Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jiwen Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jingjiang Pi
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Brian Tomlinson
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Paul Chan
- Division of CardiologyDepartment of Internal MedicineWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Qi Zhang
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Huimin Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Edward Morrisey
- Department of Cell and Developmental Biology (R.W., E.E.M.)Department of Medicine (E.E.M.)Penn Cardiovascular Institute (E.E.M.), and Penn Institute for Regenerative Medicine (E.E.M.)University of PennsylvaniaPhiladelphiaPennsylvania
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Li Lin
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina,Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina,Shanghai Jinshan Eye Disease Prevention and Treatment InstituteShanghai Jinshan Nuclear and Chemical Injury Emergency Treatment CenterJinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
8
|
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
|
9
|
Jiang Q, Wang L, Si X, Tian JL, Zhang Y, Gui HL, Li B, Tan DH. Current progress on the mechanisms of hyperhomocysteinemia-induced vascular injury and use of natural polyphenol compounds. Eur J Pharmacol 2021; 905:174168. [PMID: 33984300 DOI: 10.1016/j.ejphar.2021.174168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/09/2021] [Accepted: 05/05/2021] [Indexed: 12/28/2022]
Abstract
Cardiovascular disease is one of the most common diseases in the elderly population, and its incidence has rapidly increased with the prolongation of life expectancy. Hyperhomocysteinemia is an independent risk factor for various cardiovascular diseases, including atherosclerosis, and damage to vascular function plays an initial role in its pathogenesis. This review presents the latest knowledge on the mechanisms of vascular injury caused by hyperhomocysteinemia, including oxidative stress, endoplasmic reticulum stress, protein N-homocysteinization, and epigenetic modification, and discusses the therapeutic targets of natural polyphenols. Studies have shown that natural polyphenols in plants can reduce homocysteine levels and regulate DNA methylation by acting on oxidative stress and endoplasmic reticulum stress-related signaling pathways, thus improving hyperhomocysteinemia-induced vascular injury. Natural polyphenols obtained via daily diet are safer and have more practical significance in the prevention and treatment of chronic diseases than traditional drugs.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Lihu Road 1800, Wuxi 214122, China.
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Jin-Long Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Ye Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Hai-Long Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - De-Hong Tan
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| |
Collapse
|
10
|
Mechanisms of Endothelial Regeneration and Vascular Repair and Their Application to Regenerative Medicine. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:52-65. [PMID: 33069720 PMCID: PMC7560161 DOI: 10.1016/j.ajpath.2020.10.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Endothelial barrier integrity is required for maintaining vascular homeostasis and fluid balance between the circulation and surrounding tissues and for preventing the development of vascular disease. Despite comprehensive understanding of the molecular mechanisms and signaling pathways that mediate endothelial injury, the regulatory mechanisms responsible for endothelial regeneration and vascular repair are incompletely understood and constitute an emerging area of research. Endogenous and exogenous reparative mechanisms serve to reverse vascular damage and restore endothelial barrier function through regeneration of a functional endothelium and re-engagement of endothelial junctions. In this review, mechanisms that contribute to endothelial regeneration and vascular repair are described. Targeting these mechanisms has the potential to improve outcome in diseases that are characterized by vascular injury, such as atherosclerosis, restenosis, peripheral vascular disease, sepsis, and acute respiratory distress syndrome. Future studies to further improve current understanding of the mechanisms that control endothelial regeneration and vascular repair are also highlighted.
Collapse
|
11
|
ADMA: A Key Player in the Relationship between Vascular Dysfunction and Inflammation in Atherosclerosis. J Clin Med 2020; 9:jcm9093026. [PMID: 32962225 PMCID: PMC7563400 DOI: 10.3390/jcm9093026] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic cardiovascular disease which increases risk of major cardiovascular events including myocardial infarction and stroke. Elevated plasma concentrations of asymmetric dimethylarginine (ADMA) have long been recognised as a hallmark of cardiovascular disease and are associated with cardiovascular risk factors including hypertension, obesity and hypertriglyceridemia. In this review, we discuss the clinical literature that link ADMA concentrations to increased risk of the development of atherosclerosis. The formation of atherosclerotic lesions relies on the interplay between vascular dysfunction, leading to endothelial activation and the accumulation of inflammatory cells, particularly macrophages, within the vessel wall. Here, we review the mechanisms through which elevated ADMA contributes to endothelial dysfunction, activation and reactive oxygen species (ROS) production; how ADMA may affect vascular smooth muscle phenotype; and finally whether ADMA plays a regulatory role in the inflammatory processes occurring within the vessel wall.
Collapse
|
12
|
Zhuang T, Liu J, Chen X, Pi J, Kuang Y, Wang Y, Tomlinson B, Chan P, Zhang Q, Li Y, Yu Z, Zheng X, Reilly M, Morrisey E, Zhang L, Liu Z, Zhang Y. Cell-Specific Effects of GATA (GATA Zinc Finger Transcription Factor Family)-6 in Vascular Smooth Muscle and Endothelial Cells on Vascular Injury Neointimal Formation. Arterioscler Thromb Vasc Biol 2020; 39:888-901. [PMID: 30943773 DOI: 10.1161/atvbaha.118.312263] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objective- Transcription factor GATA (GATA zinc finger transcription factor family)-6 is highly expressed in vessels and rapidly downregulated in balloon-injured carotid arteries and viral delivery of GATA-6 to the vessels limited the neointimal formation, however, little is known about its cell-specific regulation of in vivo vascular smooth muscle cell (VSMC) phenotypic state contributing to neointimal formation. This study aims to determine the role of vascular cell-specific GATA-6 in ligation- or injury-induced neointimal hyperplasia in vivo. Approach and Results- Endothelial cell and VSMC-specific GATA-6 deletion mice are generated, and the results indicate that endothelial cell-specific GATA-6 deletion mice exhibit significant decrease of VSMC proliferation and attenuation of neointimal formation after artery ligation and injury compared with the wild-type littermate control mice. PDGF (platelet-derived growth factor)-B is identified as a direct target gene, and endothelial cell-GATA-6-PDGF-B pathway regulates VSMC proliferation and migration in a paracrine manner which controls the neointimal formation. In contrast, VSMC-specific GATA-6 deletion promotes injury-induced VSMC transformation from contractile to proliferative synthetic phenotype leading to increased neointimal formation. CCN (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed family)-5 is identified as a novel target gene, and VSMC-specific CCN-5 overexpression in mice reverses the VSMC-GATA-6 deletion-mediated increased cell proliferation and migration and finally attenuates the neointimal formation. Conclusions- This study gives us a direct in vivo evidence of GATA-6 cell lineage-specific regulation of PDGF-B and CCN-5 on VSMC phenotypic state, proliferation and migration contributing to neointimal formation, which advances our understanding of in vivo neointimal hyperplasia, meanwhile also provides opportunities for future therapeutic interventions.
Collapse
Affiliation(s)
- Tao Zhuang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jie Liu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoli Chen
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jingjiang Pi
- Department of Cardiology (Q.Z., Y.L., J.P.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yashu Kuang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yanfang Wang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Brain Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, China (B.T.)
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.)
| | - Qi Zhang
- Department of Cardiology (Q.Z., Y.L., J.P.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Ying Li
- Department of Cardiology (Q.Z., Y.L., J.P.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Zuoren Yu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, China (X.Z.).,Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, NSW, Australia (X.Z.)
| | - Muredach Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University, New York, NY (M.R.)
| | - Edward Morrisey
- Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Lin Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Zhongmin Liu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China.,Department of Cardiovascular and Thoracic Surgery (Z.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yuzhen Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., Y.K., Y.W., Z.Y., L.Z., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| |
Collapse
|
13
|
Hulin JA, Gubareva EA, Jarzebska N, Rodionov RN, Mangoni AA, Tommasi S. Inhibition of Dimethylarginine Dimethylaminohydrolase (DDAH) Enzymes as an Emerging Therapeutic Strategy to Target Angiogenesis and Vasculogenic Mimicry in Cancer. Front Oncol 2020; 9:1455. [PMID: 31993367 PMCID: PMC6962312 DOI: 10.3389/fonc.2019.01455] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The small free radical gas nitric oxide (NO) plays a key role in various physiological and pathological processes through enhancement of endothelial cell survival and proliferation. In particular, NO has emerged as a molecule of interest in carcinogenesis and tumor progression due to its crucial role in various cancer-related events including cell invasion, metastasis, and angiogenesis. The dimethylarginine dimethylaminohydrolase (DDAH) family of enzymes metabolize the endogenous nitric oxide synthase (NOS) inhibitors, asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA), and are thus key for maintaining homeostatic control of NO. Dysregulation of the DDAH/ADMA/NO pathway resulting in increased local NO availability often promotes tumor growth, angiogenesis, and vasculogenic mimicry. Recent literature has demonstrated increased DDAH expression in tumors of different origins and has also suggested a potential ADMA-independent role for DDAH enzymes in addition to their well-studied ADMA-mediated influence on NO. Inhibition of DDAH expression and/or activity in cell culture models and in vivo studies has indicated the potential therapeutic benefit of this pathway through inhibition of both angiogenesis and vasculogenic mimicry, and strategies for manipulating DDAH function in cancer are currently being actively pursued by several research groups. This review will thus provide a timely discussion on the expression, regulation, and function of DDAH enzymes in regard to angiogenesis and vasculogenic mimicry, and will offer insight into the therapeutic potential of DDAH inhibition in cancer based on preclinical studies.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ekaterina A Gubareva
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
14
|
Huang WC, Teng HI, Chen HY, Wu CJ, Tsai CT, Hsueh CH, Chen YY, Hau WK, Lu TM. Association between asymmetric dimethylarginine and in-stent restenosis tissue characteristics assessed by optical coherence tomography. Int J Cardiol 2019; 289:131-137. [DOI: 10.1016/j.ijcard.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/08/2019] [Accepted: 05/02/2019] [Indexed: 01/26/2023]
|
15
|
Farhadnejad H, Emamat H, Zand H. The Effect of Resveratrol on Cellular Senescence in Normal and Cancer Cells: Focusing on Cancer and Age-Related Diseases. Nutr Cancer 2019; 71:1175-1180. [DOI: 10.1080/01635581.2019.1597907] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hossein Farhadnejad
- Student Research Committee, Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Emamat
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Hamid Zand
- Department of Basic Medical Sciences, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Esse R, Barroso M, Tavares de Almeida I, Castro R. The Contribution of Homocysteine Metabolism Disruption to Endothelial Dysfunction: State-of-the-Art. Int J Mol Sci 2019; 20:E867. [PMID: 30781581 PMCID: PMC6412520 DOI: 10.3390/ijms20040867] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023] Open
Abstract
Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid formed during the metabolism of the essential amino acid methionine. Hcy is considered a risk factor for atherosclerosis and cardiovascular disease (CVD), but the molecular basis of these associations remains elusive. The impairment of endothelial function, a key initial event in the setting of atherosclerosis and CVD, is recurrently observed in hyperhomocysteinemia (HHcy). Various observations may explain the vascular toxicity associated with HHcy. For instance, Hcy interferes with the production of nitric oxide (NO), a gaseous master regulator of endothelial homeostasis. Moreover, Hcy deregulates the signaling pathways associated with another essential endothelial gasotransmitter: hydrogen sulfide. Hcy also mediates the loss of critical endothelial antioxidant systems and increases the intracellular concentration of reactive oxygen species (ROS) yielding oxidative stress. ROS disturb lipoprotein metabolism, contributing to the growth of atherosclerotic vascular lesions. Moreover, excess Hcy maybe be indirectly incorporated into proteins, a process referred to as protein N-homocysteinylation, inducing vascular damage. Lastly, cellular hypomethylation caused by build-up of S-adenosylhomocysteine (AdoHcy) also contributes to the molecular basis of Hcy-induced vascular toxicity, a mechanism that has merited our attention in particular. AdoHcy is the metabolic precursor of Hcy, which accumulates in the setting of HHcy and is a negative regulator of most cell methyltransferases. In this review, we examine the biosynthesis and catabolism of Hcy and critically revise recent findings linking disruption of this metabolism and endothelial dysfunction, emphasizing the impact of HHcy on endothelial cell methylation status.
Collapse
Affiliation(s)
- Ruben Esse
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Madalena Barroso
- University Children's Research@Kinder-UKE, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Isabel Tavares de Almeida
- Laboratory of Metabolism and Genetics, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
| | - Rita Castro
- Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
17
|
Madaric J, Valachovicova M, Paulis L, Pribojova J, Mateova R, Sebekova K, Postulkova L, Madaricova T, Bucova M, Mistrik M, Vulev I. Improvement in asymmetric dimethylarginine and oxidative stress in patients with limb salvage after autologous mononuclear stem cell application for critical limb ischemia. Stem Cell Res Ther 2017; 8:165. [PMID: 28697789 PMCID: PMC5506609 DOI: 10.1186/s13287-017-0622-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/19/2017] [Accepted: 06/26/2017] [Indexed: 11/10/2022] Open
Abstract
Background Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, acts as an inhibitor of angiogenesis and is associated with an increased risk of cardiovascular mortality. Administration of stem cells may affect endogenous mechanisms that regulate ADMA production and metabolism. The aim of the present study was to analyze ADMA concentration and changes in oxidative stress in patients with advanced critical limb ischemia (CLI) after bone marrow-derived mononuclear cell (BM-MNC) therapy. Methods Fifty patients (age 64 ± 11 years, 44 males, 6 females) with advanced CLI (Rutherford category 5 or 6) not eligible for revascularization were treated by intramuscular (n = 25) or intra-arterial (n = 25) injection of 40 ml BM-MNC concentrate. Patients with limb salvage and improved wound healing after 6 months were considered responders to cell therapy. The concentrations of markers of oxidative stress and angiogenesis were analyzed before, and at 3 and 6 months after BM-MNC delivery. Results At 6-month follow-up, four patients died of reasons unrelated to stem cell therapy. Among the survivors, 80% (37/46) showed limb salvage and improved wound healing. At 6 months follow-up, ADMA concentration significantly decreased in patients with limb salvage (1.74 ± 0.66 to 0.90 ± 0.49 μmol/L, p < 0.001), in parallel with decreased tumor necrosis factor (TNF)-α (2.22 ± 0.16 to 1.94 ± 0.38 pg/ml, p < 0.001), and increased reduced glutathione (6.96 ± 3.1 to 8.67 ± 4.2 μmol/L, p = 0.02), superoxide dismutase activity (168 ± 50 to 218 ± 37 U/L, p = 0.002), and coenzyme Q10 concentration (468 ± 182 to 598 ± 283 μg/L, p = 0.02). The number of delivered BM-MNCs significantly correlated with the decrease in ADMA concentration at 3 months (p = 0.004, r = −0.48) and the decrease in TNF-α concentration at 6 months (p = 0.03, r = −0.44) after cell delivery. ADMA or TNF-α improvement did not correlate with the number of applied CD34+ cells, C-reactive protein concentration, leukocyte count, or the dose of atorvastatin. Conclusions The therapeutic benefit of BM-MNC therapy is associated with reduced ADMA levels and oxidative stress. Regulation of the ADMA-nitric oxide axis and improved antioxidant status may be involved in the beneficial effects of stem cell therapy. Trial registration The study was approved and retrospectively registered by ISRCTN registry, ISRCTN16096154. Registered on 26 July 2016.
Collapse
Affiliation(s)
- Juraj Madaric
- National Institute of Cardiovascular Diseases, Slovak Medical University, Bratislava, Slovakia.
| | | | - Ludovit Paulis
- Institute of Molecular BioMedicine, Faculty of Medicine Comenius University, Bratislava, Slovakia
| | | | | | - Katarina Sebekova
- Institute of Molecular BioMedicine, Faculty of Medicine Comenius University, Bratislava, Slovakia
| | - Luba Postulkova
- National Institute of Cardiovascular Diseases, Slovak Medical University, Bratislava, Slovakia
| | - Terezia Madaricova
- National Institute of Cardiovascular Diseases, Slovak Medical University, Bratislava, Slovakia
| | - Maria Bucova
- Institute of Imunology, Faculty of Medicine Comenius University, Bratislava, Slovakia
| | - Martin Mistrik
- Clinic of Haematology and Transfusiology, Faculty Hospital, Bratislava, Slovakia
| | - Ivan Vulev
- National Institute of Cardiovascular Diseases, Slovak Medical University, Bratislava, Slovakia
| |
Collapse
|
18
|
DDAH1 plays dual roles in PM2.5 induced cell death in A549 cells. Biochim Biophys Acta Gen Subj 2016; 1860:2793-801. [DOI: 10.1016/j.bbagen.2016.03.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/04/2016] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
|
19
|
Kuang DB, Zhou JP, Yu LY, Zeng WJ, Xiao J, Zhu GZ, Zhang ZL, Chen XP. DDAH1-V3 transcript might act as miR-21 sponge to maintain balance of DDAH1-V1 in cultured HUVECs. Nitric Oxide 2016; 60:59-68. [DOI: 10.1016/j.niox.2016.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/11/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
|
20
|
Lange C, Mowat F, Sayed H, Mehad M, Duluc L, Piper S, Luhmann U, Nandi M, Kelly P, Smith A, Ali R, Leiper J, Bainbridge J. Dimethylarginine dimethylaminohydrolase-2 deficiency promotes vascular regeneration and attenuates pathological angiogenesis. Exp Eye Res 2016; 147:148-155. [PMID: 27181226 PMCID: PMC4912010 DOI: 10.1016/j.exer.2016.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 11/21/2022]
Abstract
Ischemia-induced angiogenesis is critical for tissue repair, but aberrant neovascularization in the retina causes severe sight impairment. Nitric oxide (NO) has been implicated in neovascular eye disease because of its pro-angiogenic properties in the retina. Nitric oxide production is inhibited endogenously by asymmetric dimethylarginines (ADMA and L-NMMA) which are metabolized by dimethylarginine dimethylaminohydrolase (DDAH) 1 and 2. The aim of this study was to determine the roles of DDAH1, DDAH2, ADMA and L-NMMA in retinal ischemia-induced angiogenesis. First, DDAH1, DDAH2, ADMA and L-NMMA levels were determined in adult C57BL/6J mice. The results obtained revealed that DDAH1 was twofold increased in the retina compared to the brain and the choroid. DDAH2 expression was approximately 150 fold greater in retinal and 70 fold greater in choroidal tissue compared to brain tissue suggesting an important tissue-specific role for DDAH2 in the retina and choroid. ADMA and L-NMMA levels were similar in the retina and choroid under physiological conditions. Next, characterization of DDAH1+/− and DDAH2−/− deficient mice by in vivo fluorescein angiography, immunohistochemistry and electroretinography revealed normal neurovascular function compared with wildtype control mice. Finally, DDAH1+/− and DDAH2−/− deficient mice were studied in the oxygen-induced retinopathy (OIR) model, a model used to emulate retinal ischemia and neovascularization, and VEGF and ADMA levels were quantified by ELISA and liquid chromatography tandem mass spectrometry. In the OIR model, DDAH1+/− exhibited a similar phenotype compared to wildtype controls. DDAH2 deficiency, in contrast, resulted in elevated retinal ADMA which was associated with attenuated aberrant angiogenesis and improved vascular regeneration in a VEGF independent manner. Taken together this study suggests, that in retinal ischemia, DDAH2 deficiency elevates ADMA, promotes vascular regeneration and protects against aberrant angiogenesis. Therapeutic inhibition of DDAH2 may therefore offer a potential therapeutic strategy to protect sight by promoting retinal vascular regeneration and preventing pathological angiogenesis. Nitric oxide has been implicated in neovascular eye disease. Key inhibitor of NO production is ADMA, which is metabolized by DDAH. DDAH2 deficiency results in elevated ADMA and reduced neovascularization in mice. Therapeutic inhibition of ADMA or DDAH2 may offer a potential therapeutic strategy.
Collapse
Affiliation(s)
- Clemens Lange
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK; Eye Center, University Hospital Freiburg, Germany
| | - Freya Mowat
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - Haroon Sayed
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - Manjit Mehad
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - Lucie Duluc
- The Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Sophie Piper
- The Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Ulrich Luhmann
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - Manasi Nandi
- Institute of Pharmaceutical Science, King's College London, UK
| | - Peter Kelly
- The Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Alexander Smith
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - Robin Ali
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK
| | - James Leiper
- The Nitric Oxide Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - James Bainbridge
- Department of Genetics, UCL Institute of Ophthalmology, 11-43 Bath Street, London, UK.
| |
Collapse
|
21
|
Zhao C, Li T, Han B, Yue W, Shi L, Wang H, Guo Y, Lu Z. DDAH1 deficiency promotes intracellular oxidative stress and cell apoptosis via a miR-21-dependent pathway in mouse embryonic fibroblasts. Free Radic Biol Med 2016; 92:50-60. [PMID: 26806551 DOI: 10.1016/j.freeradbiomed.2016.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 01/04/2023]
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase (NOS) inhibitor, is degraded by dimethylarginine dimethylaminohydrolase 1 (DDAH1). Emerging evidence suggests that plasma ADMA accumulation, DDAH1 activity/expression reduction, and microRNA-21 (miR-21) upregulation are linked to disease pathology, but the mechanisms remain largely unknown. In the present study, we assessed the potential role of the ADMA-DDAH1-miR-21 pathway in the regulation of the cellular redox state and apoptosis using wild-type (WT) and DDAH1-knockout (KO) immortalized mouse embryonic fibroblasts (MEFs). DDAH1 deficiency significantly increased ADMA levels, enhanced cellular oxidative stress, and rendered cells more vulnerable to apoptosis induced by tert-butyl hydroperoxide (tBHP) or A23187. However, treatment with exogenous ADMA (1-80μM) for 24h or for a prolonged period (10μM, 10 passages) in WT MEFs had no marked effect on intracellular reactive oxygen species (ROS) and apoptosis sensitivity. Interestingly, miR-21 expression was significantly increased, by 4 fold, in DDAH1(-/-) MEFs, and the induction of miR-21 by DDAH1 deficiency was dependent on oxidative stress and NF-κB activation. Inhibition of DDAH1 activity by PD 404182 also increased miR-21 expression. Furthermore, inhibition of miR-21 with a lentiviral vector in DDAH1(-/-) MEFs significantly upregulated SOD2 expression and the attenuated oxidative stress and apoptosis induced by tBHP or A23187. Taken together, our results suggest that DDAH1 not only acts as an enzyme degrading ADMA but also controls cellular oxidative stress and apoptosis via a miR-21-dependent pathway.
Collapse
Affiliation(s)
- Chenyang Zhao
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianhe Li
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingxing Han
- Key Laboratory of Space Nutrition and Food Engineering, China Astronaut Research and Training Center, Beijing 100094, China
| | - Wenhui Yue
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linlin Shi
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyun Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuting Guo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
22
|
Chertow JH, Alkaitis MS, Nardone G, Ikeda AK, Cunnington AJ, Okebe J, Ebonyi AO, Njie M, Correa S, Jayasooriya S, Casals-Pascual C, Billker O, Conway DJ, Walther M, Ackerman H. Plasmodium Infection Is Associated with Impaired Hepatic Dimethylarginine Dimethylaminohydrolase Activity and Disruption of Nitric Oxide Synthase Inhibitor/Substrate Homeostasis. PLoS Pathog 2015; 11:e1005119. [PMID: 26407009 PMCID: PMC4583463 DOI: 10.1371/journal.ppat.1005119] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/29/2015] [Indexed: 12/04/2022] Open
Abstract
Inhibition of nitric oxide (NO) signaling may contribute to pathological activation of the vascular endothelium during severe malaria infection. Dimethylarginine dimethylaminohydrolase (DDAH) regulates endothelial NO synthesis by maintaining homeostasis between asymmetric dimethylarginine (ADMA), an endogenous NO synthase (NOS) inhibitor, and arginine, the NOS substrate. We carried out a community-based case-control study of Gambian children to determine whether ADMA and arginine homeostasis is disrupted during severe or uncomplicated malaria infections. Circulating plasma levels of ADMA and arginine were determined at initial presentation and 28 days later. Plasma ADMA/arginine ratios were elevated in children with acute severe malaria compared to 28-day follow-up values and compared to children with uncomplicated malaria or healthy children (p<0.0001 for each comparison). To test the hypothesis that DDAH1 is inactivated during Plasmodium infection, we examined DDAH1 in a mouse model of severe malaria. Plasmodium berghei ANKA infection inactivated hepatic DDAH1 via a post-transcriptional mechanism as evidenced by stable mRNA transcript number, decreased DDAH1 protein concentration, decreased enzyme activity, elevated tissue ADMA, elevated ADMA/arginine ratio in plasma, and decreased whole blood nitrite concentration. Loss of hepatic DDAH1 activity and disruption of ADMA/arginine homeostasis may contribute to severe malaria pathogenesis by inhibiting NO synthesis. During a malaria infection, the vascular endothelium becomes more adhesive, permeable, and prone to trigger blood clotting. These changes help the parasite adhere to blood vessels, but endanger the host by obstructing blood flow through small vessels. Endothelial nitric oxide (NO) would normally counteract these pathological changes, but NO signalling is diminished malaria. NO synthesis is inhibited by asymmetric dimethylarginine (ADMA), a methylated derivative of arginine that is released during normal protein turnover. We found the ratio of ADMA to arginine to be elevated in Gambian children with severe malaria, a metabolic disturbance known to inhibit NO synthesis. ADMA was associated with markers of endothelial activation and impaired tissue perfusion. In parallel experiments using mice, the enzyme responsible for metabolizing ADMA, dimethylarginine dimethylaminohydrolase (DDAH), was inactivated after infection with a rodent malaria. Based on these studies, we propose that decreased metabolism of ADMA by DDAH might contribute to the elevated ADMA/arginine ratio observed during an acute episode of malaria. Strategies to preserve or increase DDAH activity might improve NO synthesis and help to prevent the vascular manifestations of severe malaria.
Collapse
Affiliation(s)
- Jessica H. Chertow
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Matthew S. Alkaitis
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington Oxford, United Kingdom
| | - Glenn Nardone
- Research Technology Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Allison K. Ikeda
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | | | | | | | | | | | | | | | - Oliver Billker
- Wellcome Trust Sanger Institute, Hinxton Cambridge, United Kingdom
| | - David J. Conway
- MRC Unit, Fajara, The Gambia
- London School of Hygiene and Tropical Medicine, Bloomsbury, London, United Kingdom
| | | | - Hans Ackerman
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Konya H, Miuchi M, Satani K, Matsutani S, Yano Y, Tsunoda T, Ikawa T, Matsuo T, Ochi F, Kusunoki Y, Tokuda M, Katsuno T, Hamaguchi T, Miyagawa JI, Namba M. Asymmetric dimethylarginine, a biomarker of cardiovascular complications in diabetes mellitus. World J Exp Med 2015; 5:110-119. [PMID: 25992325 PMCID: PMC4436934 DOI: 10.5493/wjem.v5.i2.110] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 12/23/2014] [Accepted: 02/09/2015] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular (CV) complications are an essential causal element of prospect in diabetes mellitus (DM), with carotid atherosclerosis being a common risk factor for prospective crisis of coronary artery diseases and/or cerebral infarction in DM subjects. From another point of view, asymmetric dimethylarginine (ADMA) has been established as an inhibitor of endogenous nitric oxide synthesis and the relationship between ADMA and arteriosclerosis has been reported. In our study with 87 type 2 DM (T2DM) patients, we have examined whether ADMA and other CV risk factors are the useful predictors of DMCV complications. After the measurement of the respective CV risk factors, we have followed the enrolled T2DM patients for 5 years. We have finally analyzed 77 patients. DMCV complications developed in 15 cases newly within 5 years, and 4 cases recurred. The concentrations of ADMA in plasma were markedly more elevated in 19 DM patients with CV complications than in 58 DM patients without CV complications. Urinary albumin (U-Alb), mean intimal-medial thickness (IMT) and ankle brachial index (ABI) were also higher in patients with CV complications. Multiple regression analyses showed that U-Alb had an influence on the high level of ADMA (standardized β = 6.59, P = 0.00014) independently of age, systolic BP, fibrinogen, mean IMT, plaque score, and ABI. The review indicates what is presently known regarding plasma ADMA that might be a new and meaningful biomarker of CV complications in DM subjects.
Collapse
|
24
|
Zhang F, Li X, Dong Q, Wang Y, Zhang H. Risk of Acute Cerebral Infarction and Plasma Asymmetrical Dimethylarginine and Homocysteine Levels: A Clinical Correlation Analysis of Chinese Population. J Stroke Cerebrovasc Dis 2014; 23:2225-32. [DOI: 10.1016/j.jstrokecerebrovasdis.2014.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 03/27/2014] [Accepted: 04/02/2014] [Indexed: 11/29/2022] Open
|
25
|
The endothelial ADMA/NO pathway in hypoxia-related chronic respiratory diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:501612. [PMID: 24719871 PMCID: PMC3955646 DOI: 10.1155/2014/501612] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/18/2014] [Indexed: 01/20/2023]
Abstract
Since its discovery, many adhere to the view that asymmetric dimethylarginine (ADMA), as an inhibitor of the synthesis of nitric oxide (NO), contributes to the pathogenesis of various diseases. Particularly, this is evident in disease of the cardiovascular system, in which endothelial dysfunction results in an imbalance between vasoconstriction and vasodilatation. Even if increased ADMA concentrations are closely related to an endothelial dysfunction, several studies pointed to a potential beneficial effect of ADMA, mainly in the context of angioproliferative disease such as cancer and fibrosis. Antiproliferative properties of ADMA independent of NO have been identified in this context. In particular, the regulation of ADMA by its degrading enzyme dimethylarginine dimethylaminohydrolase (DDAH) is the object of many studies. DDAH is discussed as a promising therapeutic target for the indirect regulation of NO. In hypoxia-related chronic respiratory diseases, this controversy discussion of ADMA and DDAH is particularly evident and is therefore subject of this review.
Collapse
|
26
|
Klinaki E, Soldatou A, Marmarinos A, Pagoni A, Tsentidis C, Gourgiotis D, Garoufi A. The Role of Asymmetric Dimethylarginine and Lipoprotein Associated Phospholipase A2 in Children and Adolescents with Dyslipidemia. Health (London) 2014. [DOI: 10.4236/health.2014.612177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Lin IC, Sheen JM, Tain YL, Chou MH, Huang LT, Yang KD. Vascular Endothelial Growth Factor-A in <i>Lactobacillus Casei </i>Cell Wall Extract-Induced Coronary Arteritis of a Murine Model. Circ J 2014; 78:752-762. [DOI: 10.1253/circj.cj-13-0612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Ming-Huei Chou
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine
| | - Kuender D. Yang
- Department of Medical Research, Show Chwan Memorial Hospital in Chang Bing
| |
Collapse
|
28
|
Ghebremariam YT, Huang NF, Kambhampati S, Volz KS, Joshi GG, Anslyn EV, Cooke JP. Characterization of a fluorescent probe for imaging nitric oxide. J Vasc Res 2013; 51:68-79. [PMID: 24335468 PMCID: PMC3927988 DOI: 10.1159/000356445] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 10/11/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Nitric oxide (NO), a potent vasodilator and anti-atherogenic molecule, is synthesized in various cell types, including vascular endothelial cells (ECs). The biological importance of NO enforces the need to develop and characterize specific and sensitive probes. To date, several fluorophores, chromophores and colorimetric techniques have been developed to detect NO or its metabolites (NO(2) and NO(3)) in biological fluids, viable cells or cell lysates. METHODS Recently, a novel probe (NO(550)) has been developed and reported to detect NO in solutions and in primary astrocytes and neuronal cells with a fluorescence signal arising from a nonfluorescent background. RESULTS Here, we report further characterization of this probe by optimizing conditions for the detection and imaging of NO products in primary vascular ECs, fibroblasts, and embryonic stem cell- and induced pluripotent stem cell-derived ECs in the absence and presence of pharmacological agents that modulate NO levels. In addition, we studied the stability of this probe in cells over time and evaluated its compartmentalization in reference to organelle-labeling dyes. Finally, we synthesized an inherently fluorescent diazo ring compound (AZO(550)) that is expected to form when the nonfluorescent NO(550) reacts with cellular NO, and compared its cellular distribution with that of NO(550). CONCLUSION NO(550) is a promising agent for imaging NO at baseline and in response to pharmacological agents that modulate its levels.
Collapse
Affiliation(s)
- Yohannes T Ghebremariam
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ngan F Huang
- Stanford cardiovascular Institute, Stanford, California 94305, USA
| | | | - Katharina S Volz
- Stanford cardiovascular Institute, Stanford, California 94305, USA
| | - Gururaj G Joshi
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712, USA
| | - Eric V Anslyn
- Department of Chemistry and Biochemistry, University of Texas, Austin, Texas 78712, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
29
|
Sun T, Zhou JP, Kuang DB, Li MP, Xiong Y, Tang J, Xia J, Bai YP, Yang GP, Li YJ, Chen XP. Correlations of DDAH1 transcript variants with human endothelial asymmetric dimethylarginine metabolizing activity. Am J Hypertens 2013; 26:1437-44. [PMID: 23864585 DOI: 10.1093/ajh/hpt119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Dimethylarginine dimethylaminohydrolases 1 (DDAH1) is the major enzyme responsible for inactivation of asymmetric dimethylarginine (ADMA). This study seeks to clarify the correlations between mRNA expression levels of DDAH1 transcript variants and the relationship with ADMA metabolizing activity in human. METHODS The mRNA expression levels of DDAH1 transcript variants in primarily cultured human umbilical vein endothelial cells (HUVECs) and peripheral blood mononuclear cells (PBMCs) from healthy control subjects and patients suffering from both acute ischemic stroke (AIS) and acute myocardial infarction (AMI) were determined by real-time polymerase chain reaction. ADMA metabolizing activity of the cell lysates from HUVECs was determined by enzyme-linked immunosorbent assay. RESULTS A novel DDAH1 transcript variant DDAH1-V3 was identified. DDAH1-V3 mRNA expression correlated significantly with that of both -V2 (R = 0.811; P = 0.000008) and -V1 (R = 0.454; P = 0.04) in HUVECs. In PBMCs from healthy subjects, significant correlation was observed only between DDAH1-V2 and -V3 (R = 0.571; P = 0.001; n = 36). Delta threshold cycle (DCT) values for both DDAH1-V2 and -V3 transcripts were increased significantly in PBMCs from AIS patients (P < 0.05, respectively). In PBMCs from patients suffering from both AIS and AMI, positive pairwise correlations between mRNA levels of DDAH1 transcripts were also observed as analyzed by partial correlation analysis (P < 0.05, respectively). However, only mRNA expression level of the DDAH1-V1 transcript correlated significantly with intracellular ADMA metabolizing activity in HUVECs (R = 0.805; P=0.002). CONCLUSIONS This study demonstrated that although there are positive correlations between mRNA expression levels of DDAH1 transcript variants, only the DDAH1-V1 transcript is responsible for ADMA metabolism, and transcript specific primers are recommended to determine DDAH1 mRNA expression.
Collapse
Affiliation(s)
- Tao Sun
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University; Changsha, Hunan, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhang P, Xu X, Hu X, Wang H, Fassett J, Huo Y, Chen Y, Bache RJ. DDAH1 deficiency attenuates endothelial cell cycle progression and angiogenesis. PLoS One 2013; 8:e79444. [PMID: 24260221 PMCID: PMC3832548 DOI: 10.1371/journal.pone.0079444] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/23/2013] [Indexed: 11/18/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide (NO) synthase (NOS). ADMA is eliminated largely by the action of dimethylarginine dimethylaminohydrolase1 (DDAH1). Decreased DDAH activity is found in several pathological conditions and is associated with increased risk of vascular disease. Overexpression of DDAH1 has been shown to augment endothelial proliferation and angiogenesis. To better understand the mechanism by which DDAH1 influences endothelial proliferation, this study examined the effect of DDAH1 deficiency on cell cycle progression and the expression of some cell cycle master regulatory proteins. DDAH1 KO decreased in vivo Matrigel angiogenesis and depressed endothelial repair in a mouse model of carotid artery wire injury. DDAH1 deficiency decreased VEGF expression in HUVEC and increased NF1 expression in both HUVEC and DDAH1 KO mice. The expression of active Ras could overcome the decreased VEGF expression caused by the DDAH1 depletion. The addition of VEGF and knockdown NF1 could both restore proliferation in cells with DDAH1 depletion. Flow cytometry analysis revealed that DDAH1 sRNAi knockdown in HUVEC caused G1 and G2/M arrest that was associated with decreased expression of CDC2, CDC25C, cyclin D1 and cyclin E. MEF cells from DDAH1 KO mice also demonstrated G2/M arrest that was associated with decreased cyclin D1 expression and Akt activity. Our findings indicate that DDAH1 exerts effects on cyclin D1 and cyclin E expression through multiple mechanisms, including VEGF, the NO/cGMP/PKG pathway, the Ras/PI3K/Akt pathway, and NF1 expression. Loss of DDAH1 effects on these pathways results in impaired endothelial cell proliferation and decreased angiogenesis. The findings provide background information that may be useful in the development of therapeutic strategies to manipulate DDAH1 expression in cardiovascular diseases or tumor angiogenesis.
Collapse
Affiliation(s)
- Ping Zhang
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Xin Xu
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Xinli Hu
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Huan Wang
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - John Fassett
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Yingjie Chen
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Robert J. Bache
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| |
Collapse
|
31
|
Zhang Y, Lv Y, Liu YJ, Yang C, Hu HJ, Meng XE, Li MX, Pan SY. Hyperbaric oxygen therapy in rats attenuates ischemia-reperfusion testicular injury through blockade of oxidative stress, suppression of inflammation, and reduction of nitric oxide formation. Urology 2013; 82:489.e9-489.e15. [PMID: 23769121 DOI: 10.1016/j.urology.2013.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/22/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To evaluate the therapeutic utility of hyperbaric oxygen (HBO) therapy on testicular ischemia/reperfusion (I/R) injury and elucidate the underlying molecular mechanism, we tested whether HBO therapy provided rescue of the testes after torsion in rats. METHODS Sprague-Dawley rats were randomly divided into 4 groups: control group, control plus HBO therapy, I/R group, and I/R plus HBO therapy. The I/R model was induced by torsion of the right testis. RESULTS I/R in the testis resulted in disrupted seminiferous tubules, germ cell-specific apoptosis, followed by a marked reduction in testis weight and daily sperm production. HBO therapy preserved seminiferous tubules, suppressed apoptosis, and prevented testicular atrophy in I/R testes. HBO therapy abated oxidative stress in I/R testes, marked by reduced malondialdehyde formation, enhanced activities of superoxide dismutase and heme oxygenase 1 (HO-1), and decreased activities of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and xanthine oxidase. HBO therapy resulted in a reduction of myeloperoxidase (MPO) activity in I/R testes, a marker of neutrophil recruitment. HBO therapy suppressed inflammation in I/R testes, marked by reduced messenger RNA (mRNA) levels of tumor necrosis factor-α (TNF-α), interleukin-1beta (IL-1β), and CD44. Furthermore, HBO therapy suppressed the activation of nuclear factor kappa B (NFκB), p38, and c-JUN-N-terminal kinase (JNK) signaling pathways in I/R testes. In addition, HBO therapy reduced nitric oxide formation in I/R testes through suppression of inducible nitric oxide synthase and dimethylarginine dimethylaminohydrolase. CONCLUSION HBO therapy in rats attenuated I/R-induced testicular injury, possibly through abating oxidative stress, suppressing inflammation, and reducing nitric oxide formation.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Hyperbaric Oxygen, PLA Navy General Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Chen S, Li N, Deb-Chatterji M, Dong Q, Kielstein JT, Weissenborn K, Worthmann H. Asymmetric dimethyarginine as marker and mediator in ischemic stroke. Int J Mol Sci 2012; 13:15983-6004. [PMID: 23443106 PMCID: PMC3546674 DOI: 10.3390/ijms131215983] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/14/2012] [Accepted: 11/21/2012] [Indexed: 02/07/2023] Open
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide synthase (NOS) inhibitor, is known as mediator of endothelial cell dysfunction and atherosclerosis. Circulating ADMA levels are correlated with cardiovascular risk factors such as hypercholesterolemia, arterial hypertension, diabetes mellitus, hyperhomocysteinemia, age and smoking. Accordingly, clinical studies found evidence that increased ADMA levels are associated with a higher risk of cerebrovascular events. After the acute event of ischemic stroke, levels of ADMA and its analog symmetric dimethylarginine (SDMA) are elevated through augmentation of protein methylation and oxidative stress. Furthermore, cleavage of ADMA through dimethylarginine dimethylaminohydrolases (DDAHs) is reduced. This increase of dimethylarginines might be predictive for adverse clinical outcome. However, the definite role of ADMA after acute ischemic stroke still needs to be clarified. On the one hand, ADMA might contribute to brain injury by reduction of cerebral blood flow. On the other hand, ADMA might be involved in NOS-induced oxidative stress and excitotoxic neuronal death. In the present review, we highlight the current knowledge from clinical and experimental studies on ADMA and its role for stroke risk and ischemic brain injury in the hyperacute stage after stroke. Finally, further studies are warranted to unravel the relevance of the close association of dimethylarginines with stroke.
Collapse
Affiliation(s)
- Shufen Chen
- Department of Neurology, Hannover Medical School, 30623 Hannover, Germany; E-Mails: (S.C.); (N.L.); (M.D.-C.); (K.W.)
- Department of Neurology, Huashan Hospital Fudan University, Shanghai 200040, China; E-Mail:
| | - Na Li
- Department of Neurology, Hannover Medical School, 30623 Hannover, Germany; E-Mails: (S.C.); (N.L.); (M.D.-C.); (K.W.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 10050, China
| | - Milani Deb-Chatterji
- Department of Neurology, Hannover Medical School, 30623 Hannover, Germany; E-Mails: (S.C.); (N.L.); (M.D.-C.); (K.W.)
| | - Qiang Dong
- Department of Neurology, Huashan Hospital Fudan University, Shanghai 200040, China; E-Mail:
| | - Jan T. Kielstein
- Department of Nephrology and Hypertension, Hannover Medical School, 30623 Hannover, Germany; E-Mail:
| | - Karin Weissenborn
- Department of Neurology, Hannover Medical School, 30623 Hannover, Germany; E-Mails: (S.C.); (N.L.); (M.D.-C.); (K.W.)
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Hans Worthmann
- Department of Neurology, Hannover Medical School, 30623 Hannover, Germany; E-Mails: (S.C.); (N.L.); (M.D.-C.); (K.W.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-511-532-3580; Fax: +49-511-532-3115
| |
Collapse
|
33
|
Altmann KS, Havemeyer A, Beitz E, Clement B. Dimethylarginine-dimethylaminohydrolase-2 (DDAH-2) does not metabolize methylarginines. Chembiochem 2012; 13:2599-604. [PMID: 23125090 DOI: 10.1002/cbic.201200499] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Indexed: 01/18/2023]
Abstract
Free endogenous methylarginines, N(ω)-monomethyl-L-arginine (L-NMMA) and N(ω),N(ω')-dimethyl-L-arginine (ADMA), inhibit NO synthases (NOSs) and are metabolized by dimethylargininedimethylaminohydrolase (DDAH). A postulated metabolism has been shown several times for DDAH-1, but the involvement of DDAH-2 in the degradation of ADMA and L-NMMA is still a matter of debate. Determination of the isoform-specific DDAH protein expression profiles in various porcine tissue types shows a correlation of DDAH activity only with DDAH-1 levels. DDAH activity (measured as L-citrulline formation from the conversion of methylarginines and alternative DDAH substrates) was detected in DDAH-1-rich porcine tissue types, that is, kidney, liver, and brain, but not in DDAH-2-rich porcine fractions, that is, spleen and thyroid. Furthermore, several ex vivo studies showed DDAH activity to be important for L-citrulline formation in porcine tissue and indicated the absence of an endogenous DDAH inhibitor in porcine tissue. This study provides new insights into tissue distributions as well as substrate selectivity for both DDAH isoforms. Although DDAH-1 is known to metabolize the endogenous NOS inhibitors L-NMMA and ADMA, a physiological function for DDAH-2 has yet to be determined. Hence, determining DDAH activity by methylarginine conversion is not suitable for analyzing isoform selectivity of DDAH-1 inhibitors as postulated.
Collapse
Affiliation(s)
- Karin S Altmann
- Department of Pharmaceutical and Medicinal Chemistry, Pharmaceutical Institute, Christian-Albrechts-Universität Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | | | | | | |
Collapse
|
34
|
Dimethylarginine dimethylaminohydrolase1 is an organ-specific mediator of end organ damage in a murine model of hypertension. PLoS One 2012; 7:e48150. [PMID: 23110194 PMCID: PMC3482201 DOI: 10.1371/journal.pone.0048150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 09/21/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) is an independent predictor of cardiovascular and overall mortality. Moreover, elevated ADMA plasma concentrations are associated with the extent of hypertension. However, data from small-sized clinical trials and experimental approaches using murine transgenic models have revealed conflicting results regarding the impact of ADMA and its metabolizing enzyme dimethylarginine dimethylaminohydrolase (DDAH) in the pathogenesis of hypertension. METHODOLOGY/PRINCIPAL FINDINGS Therefore, we investigated the role of ADMA and DDAH1 in hypertension-induced end organ damage using the uninephrectomized, deoxycorticosterone actetate salt, and angiotensin II-induced hypertension model in human DDAH1 (hDDAH1) overexpressing and wild-type (WT) mice. ADMA plasma concentrations differed significantly between hDDAH1 and WT mice at baseline, but did not significantly change during the induction of hypertension. hDDAH1 overexpression did not protect against hypertension-induced cardiac fibrosis and hypertrophy. In addition, the hypertension-induced impairment of the endothelium-dependent vasorelaxation of aortic segments ex vivo was not significantly attenuated by hDDAH1 overexpression. However, hDDAH1 mice displayed an attenuated hypertensive inflammatory response in renal tissue, resulting in less hypertensive renal injury. CONCLUSION/SIGNIFICANCE Our data reveal that hDDAH1 organ-specifically modulates the inflammatory response in this murine model of hypertension. The lack of protection in cardiac and aortic tissues may be due to DDAH1 tissue selectivity and/or the extent of hypertension by the used combined model. However, our study underlines the potency of hDDAH1 overexpression in modulating inflammatory processes as a crucial step in the pathogenesis of hypertension, which needs further experimental and clinical investigation.
Collapse
|
35
|
Lu TM, Chung MY, Lin MW, Hsu CP, Lin SJ. Plasma asymmetric dimethylarginine predicts death and major adverse cardiovascular events in individuals referred for coronary angiography. Int J Cardiol 2011; 153:135-40. [DOI: 10.1016/j.ijcard.2011.06.120] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 05/27/2011] [Accepted: 06/25/2011] [Indexed: 11/28/2022]
|
36
|
Synergistic adverse prognostic effects of asymmetric dimethylarginine and endothelial progenitor-related cells deficiency after elective coronary angioplasty. Int J Cardiol 2011; 152:400-3. [DOI: 10.1016/j.ijcard.2011.08.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 08/17/2011] [Indexed: 11/22/2022]
|
37
|
Boult JK, Walker-Samuel S, Jamin Y, Leiper JM, Whitley GSJ, Robinson SP. Active site mutant dimethylarginine dimethylaminohydrolase 1 expression confers an intermediate tumour phenotype in C6 gliomas. J Pathol 2011; 225:344-52. [PMID: 21590769 DOI: 10.1002/path.2904] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 02/16/2011] [Accepted: 03/17/2011] [Indexed: 12/11/2022]
Abstract
Dimethylarginine dimethylaminohydrolase (DDAH) metabolizes the endogenous inhibitor of nitric oxide synthesis, asymmetric dimethylarginine (ADMA). Constitutive over-expression of DDAH1, the isoform primarily associated with neuronal nitric oxide synthase (nNOS) results in increased tumour growth and vascularization, and elevated VEGF secretion. To address whether DDAH1-mediated tumour growth is reliant upon the enzymatic activity of DDAH1, cell lines expressing an active site mutant of DDAH1 incapable of metabolizing ADMA were created. Xenografts derived from these cell lines grew significantly faster than those derived from control cells, yet not as fast as those over-expressing wild-type DDAH1. VEGF expression in DDAH1 mutant-expressing tumours did not differ from control tumours but was significantly lower than that of wild-type DDAH1-over-expressing tumours. Fluorescence microscopy for CD31 and pimonidazole adduct formation demonstrated that DDAH1 mutant-expressing tumours had a lower endothelial content and demonstrated less hypoxia, respectively, than wild-type DDAH1-expressing tumours. However, there was no difference in uptake of the perfusion marker Hoechst 33342. Non-invasive multiparametric quantitative MRI, including the measurement of native T(1) and T(2) relaxation times and apparent water diffusion coefficient, was indicative of higher cellularity in DDAH1-expressing xenografts, which was confirmed by histological quantification of necrosis. C6 xenografts expressing active site mutant DDAH1 displayed an intermediate phenotype between tumours over-expressing wild-type DDAH1 and control tumours. These data suggest that enhanced VEGF expression downstream of DDAH1 was dependent upon ADMA metabolism, but that the DDAH1-mediated increase in tumour growth was only partially dependent upon its enzymatic activity, and therefore must involve an as-yet unidentified mechanism. DDAH1 is an important mediator of tumour progression, but appears to have addition roles independent of its metabolism of ADMA, which need to be considered in therapeutic strategies targeted against the NO/DDAH pathway in cancer.
Collapse
Affiliation(s)
- Jessica Kr Boult
- CRUK and EPSRC Cancer Imaging Centre, The Institute of Cancer Research and Royal Marsden NHS Trust, Sutton, Surrey, UK
| | | | | | | | | | | |
Collapse
|
38
|
Sibal L, Agarwal SC, Home PD, Boger RH. The Role of Asymmetric Dimethylarginine (ADMA) in Endothelial Dysfunction and Cardiovascular Disease. Curr Cardiol Rev 2011; 6:82-90. [PMID: 21532773 PMCID: PMC2892080 DOI: 10.2174/157340310791162659] [Citation(s) in RCA: 334] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Revised: 10/14/2009] [Accepted: 10/25/2009] [Indexed: 02/07/2023] Open
Abstract
Endothelium plays a crucial role in the maintenance of vascular tone and structure. Endothelial dysfunction is known to precede overt coronary artery disease. A number of cardiovascular risk factors, as well as metabolic diseases and systemic or local inflammation cause endothelial dysfunction. Nitric oxide (NO) is one of the major endothelium derived vaso-active substances whose role is of prime importance in maintaining endothelial homeostasis. Low levels of NO are associated with impaired endothelial function. Asymmetric dimethylarginine (ADMA), an analogue of L-arginine, is a naturally occurring product of metabolism found in human circulation. Elevated levels of ADMA inhibit NO synthesis and therefore impair endothelial function and thus promote atherosclerosis. ADMA levels are increased in people with hypercholesterolemia, atherosclerosis, hypertension, chronic heart failure, diabetes mellitus and chronic renal failure. A number of studies have reported ADMA as a novel risk marker of cardiovascular disease. Increased levels of ADMA have been shown to be the strongest risk predictor, beyond traditional risk factors, of cardiovascular events and all-cause and cardiovascular mortality in people with coronary artery disease. Interventions such as treatment with L-arginine have been shown to improve endothelium-mediated vasodilatation in people with high ADMA levels. However the clinical utility of modifying circulating ADMA levels remains uncertain.
Collapse
Affiliation(s)
- Latika Sibal
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | |
Collapse
|
39
|
|
40
|
Pullamsetti SS, Savai R, Dumitrascu R, Dahal BK, Wilhelm J, Konigshoff M, Zakrzewicz D, Ghofrani HA, Weissmann N, Eickelberg O, Guenther A, Leiper J, Seeger W, Grimminger F, Schermuly RT. The role of dimethylarginine dimethylaminohydrolase in idiopathic pulmonary fibrosis. Sci Transl Med 2011; 3:87ra53. [PMID: 21677199 DOI: 10.1126/scitranslmed.3001725] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, dysregulated response to alveolar injury that culminates in compromised lung function from excess extracellular matrix production. Associated with high morbidity and mortality, IPF is generally refractory to current pharmacological therapies. We examined fibrotic lungs from mice and from patients with IPF and detected increased expression of dimethylarginine dimethylaminohydrolases (DDAHs)--key enzymes that metabolize asymmetric dimethylarginine (ADMA), which is an endogenous inhibitor of nitric oxide synthase, to form l-citrulline and dimethylamine. DDAHs are up-regulated in primary alveolar epithelial type II cells from these mice and patients where they are colocalized with inducible nitric oxide synthase. In cultured alveolar epithelial type II cells from bleomycin-induced fibrotic mouse lungs, inhibition of DDAH suppressed proliferation and induced apoptosis in an ADMA-dependent manner. In addition, DDAH inhibition reduced collagen production by fibroblasts in an ADMA-independent but transforming growth factor/SMAD-dependent manner. In mice with bleomycin-induced pulmonary fibrosis, the DDAH inhibitor L-291 reduced collagen deposition and normalized lung function. In bleomycin-induced fibrosis, inducible nitric oxide synthase inhibition decreased fibrosis, but an even stronger reduction was observed after inhibition of DDAH. Thus, DDAH inhibition reduces fibroblast-induced collagen deposition in an ADMA-independent manner and reduces abnormal epithelial proliferation in an ADMA-dependent manner, offering a possible therapeutic avenue for attenuation of pulmonary fibrosis.
Collapse
|
41
|
Tuygun AK, Tuygun A, Yurtseven N, Şensöz Y, Günay R, Keser M, Tuygun UH, Ketenci B, Şahin S, Yekeler I. Asymmetric Dimethylarginine Levels in Buerger’s Disease. Ann Vasc Surg 2011; 25:547-54. [DOI: 10.1016/j.avsg.2010.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 12/10/2010] [Accepted: 12/12/2010] [Indexed: 10/18/2022]
|
42
|
Li X, Tse H, Jin L. Novel Endothelial Biomarkers: Implications for Periodontal Disease and CVD. J Dent Res 2011; 90:1062-9. [DOI: 10.1177/0022034510397194] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Endothelial cells are actively involved in various aspects of vascular biology and different stages of atherosclerosis. Endothelial function is increasingly used as an important outcome measure in cardiovascular research. Endothelial progenitor cells (EPCs) are closely linked to endothelial function, and their biomarkers have received much attention. EPCs may not only serve as a pool of progenitor cells and possess the capacity to repair the damaged vasculature, but also act as potent effectors in systemic inflammation, suggesting that EPCs may play a critical role in maintaining endothelial function and the progression of cardiovascular disease (CVD). Emerging evidence shows an association of periodontal infections (gingivitis and periodontitis) with endothelial dysfunction, while the relevant mechanisms remain unknown. Our recent finding of the association of periodontitis with EPCs warrants their utilization as additional biomarkers in future studies on periodontal medicine. This review starts with a brief account on the current understanding of the nature of periodontal infections and their link with systemic inflammation and endothelial dysfunction. The paper also provides an update on endothelial biology and function as well as the novel biomarkers of EPCs and concludes with clinical studies on periodontal diseases and CVD.
Collapse
Affiliation(s)
- X. Li
- Faculty of Dentistry, Periodontology, The University of Hong Kong, 34
Hospital Road
| | - H.F. Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - L.J. Jin
- Faculty of Dentistry, Periodontology, The University of Hong Kong, 34
Hospital Road
| |
Collapse
|
43
|
Thum T, Schmitter K, Fleissner F, Wiebking V, Dietrich B, Widder JD, Jazbutyte V, Hahner S, Ertl G, Bauersachs J. Impairment of endothelial progenitor cell function and vascularization capacity by aldosterone in mice and humans. Eur Heart J 2010; 32:1275-86. [PMID: 20926363 PMCID: PMC3094546 DOI: 10.1093/eurheartj/ehq254] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aims Hyperaldosteronism is associated with vascular injury and increased cardiovascular events. Bone marrow-derived endothelial progenitor cells (EPCs) play an important role in endothelial repair and vascular homeostasis. We hypothesized that hyperaldosteronism impairs EPC function and vascularization capacity in mice and humans. Methods and results We characterized the effects of aldosterone and mineralocorticoid receptor (MR) blockade on EPC number and function as well as vascularization capacity and endothelial function. Treatment of human EPC with aldosterone induced translocation of the MR and impaired multiple cellular functions of EPC, such as differentiation, migration, and proliferation in vitro. Impaired EPC function was rescued by pharmacological blockade or genetic ablation of the MR. Aldosterone protein kinase A (PKA) dependently increased reactive oxygen species formation in EPC. Aldosterone infusion in mice impaired EPC function, EPC homing to vascular structures and vascularization capacity in a MR-dependent but blood pressure-independent manner. Endothelial progenitor cells from patients with primary hyperaldosteronism compared with controls of similar age displayed reduced migratory potential. Impaired EPC function was associated with endothelial dysfunction. MR blockade in patients with hyperaldosteronism improved EPC function and arterial stiffness. Conclusion Endothelial progenitor cells express a MR that mediates functional impairment by PKA-dependent increase of reactive oxygen species. Normalization of EPC function may represent a novel mechanism contributing to the beneficial effects of MR blockade in cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Thomas Thum
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
- Corresponding author. Tel: +49 511 532 5272 (T.T), +49 511 532 3840 (J.B.); Fax: +49 511 532 5274 (T.T.), +49 511 532 5412 (J.B.), (T.T.), (J.B.)
| | - Kerstin Schmitter
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Felix Fleissner
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
| | - Volker Wiebking
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Bernd Dietrich
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Julian D. Widder
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Virginija Jazbutyte
- Institut für Molekulare und Translationale Therapiestrategien, Medizinische Hochschule Hannover, Germany
| | - Stefanie Hahner
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Georg Ertl
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
| | - Johann Bauersachs
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Germany
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Germany
- Corresponding author. Tel: +49 511 532 5272 (T.T), +49 511 532 3840 (J.B.); Fax: +49 511 532 5274 (T.T.), +49 511 532 5412 (J.B.), (T.T.), (J.B.)
| |
Collapse
|
44
|
Wilson AM, Shin DS, Weatherby C, Harada RK, Ng MK, Nair N, Kielstein J, Cooke JP. Asymmetric dimethylarginine correlates with measures of disease severity, major adverse cardiovascular events and all-cause mortality in patients with peripheral arterial disease. Vasc Med 2010; 15:267-74. [PMID: 20484311 PMCID: PMC3131178 DOI: 10.1177/1358863x10364552] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Peripheral arterial disease (PAD) is associated with major cardiovascular morbidity and mortality. Abnormalities in nitric oxide metabolism due to excess of the NO synthase inhibitor asymmetric dimethylarginine (ADMA) may be pathogenic in PAD. We explored the association between ADMA levels and markers of atherosclerosis, function, and prognosis. A total of 133 patients with symptomatic PAD were enrolled. Ankle-brachial index (ABI), walking time, vascular function measures (arterial compliance and flow-mediated vasodilatation) and plasma ADMA level were assessed for each patient at baseline. ADMA correlated inversely with ABI (r = -0.238, p = 0.003) and walking time (r = -0.255, p = 0.001), independent of other vascular risk factors. We followed up 125 (94%) of our 133 initial subjects with baseline measurements (mean 35 months). Subjects with ADMA levels in the highest quartile (> 0.84 mumol/l) showed a significantly greater occurrence of a major adverse cardiovascular event (MACE) compared to those with ADMA levels in the lower three quartiles (p = 0.001). Cox proportional-hazards regression analysis revealed that ADMA was a significant predictor of MACE, independent of other risk factors including age, sex, blood pressure, smoking history, diabetes and ABI (hazard ratio = 5.1, p < 0.001). Measures of vascular function, such as compliance, flow-mediated vasodilatation (FMVD) and blood pressure, as well as markers of PAD severity, including ABI and walking time, were not predictive. In conclusion, circulating levels of ADMA correlate independently with measures of disease severity and major adverse cardiovascular events. Agents that target this pathway may be useful for this patient population. Clinical Trial Registration - URL: http:// www.clinicaltrials.gov. Unique identifier: NCT00284076.
Collapse
Affiliation(s)
- Andrew M Wilson
- Falk Cardiovascular Research Institute, Stanford University Medical Center, Stanford, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Yuan Q, Peng J, Liu SY, Wang CJ, Xiang DX, Xiong XM, Hu CP, Li YJ. Inhibitory effect of resveratrol derivative BTM-0512 on high glucose-induced cell senescence involves dimethylaminohydrolase/asymmetric dimethylarginine pathway. Clin Exp Pharmacol Physiol 2010; 37:630-5. [DOI: 10.1111/j.1440-1681.2010.05368.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Jacobi J, Maas R, Cardounel AJ, Arend M, Pope AJ, Cordasic N, Heusinger-Ribeiro J, Atzler D, Strobel J, Schwedhelm E, Böger RH, Hilgers KF. Dimethylarginine dimethylaminohydrolase overexpression ameliorates atherosclerosis in apolipoprotein E-deficient mice by lowering asymmetric dimethylarginine. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2559-70. [PMID: 20348244 DOI: 10.2353/ajpath.2010.090614] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, is increasingly recognized as a novel biomarker in cardiovascular disease. To date, it remains unclear whether elevated ADMA levels are merely associated with cardiovascular risk or whether this molecule is of functional relevance in the pathogenesis of atherosclerotic vascular disease. To clarify this issue, we crossed dimethylarginine dimethylaminohydrolase (DDAH) transgenic mice that overexpress the human isoform 1 of the ADMA degrading enzyme DDAH into ApoE-deficient mice to generate ApoE(-/-)/hDDAH1(+/-) mice. In these mice, as well as ApoE(-/-) wild-type littermates, atherosclerosis within the aorta as well as vascular function of aortic ring preparations was assessed. We report here that overexpression of hDDAH1 reduces plaque formation in ApoE(-/-) mice by lowering ADMA. The extent of atherosclerosis closely correlated with plasma ADMA levels in male but not female mice fed either a standard rodent chow or an atherogenic diet. Functional analysis of aortic ring preparations revealed improved endothelial function in mice overexpressing hDDAH1. Our findings provide proof-of-principle that ADMA plays a causal role as a culprit molecule in atherosclerosis and support recent evidence indicating a functional relevance of DDAH enzymes in genetic mouse models. Together, these results demonstrate that pharmacological interventions targeting the ADMA/DDAH pathway may represent a novel approach in the prevention and management of cardiovascular diseases.
Collapse
Affiliation(s)
- Johannes Jacobi
- Department of Nephrology and Hypertension, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Schwedhelm E, von Leitner EC, Atzler D, Schmitz C, Jacobi J, Meinertz T, Münzel T, Baldus S, Cooke JP, Böger RH, Maas R, Sydow K. Extensive characterization of the human DDAH1 transgenic mice. Pharmacol Res 2009; 60:494-502. [DOI: 10.1016/j.phrs.2009.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 07/31/2009] [Accepted: 08/01/2009] [Indexed: 11/16/2022]
|
48
|
The DDAH/ADMA pathway in the control of endothelial cell migration and angiogenesis. Biochem Soc Trans 2009; 37:1243-7. [DOI: 10.1042/bst0371243] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
ADMA (asymmetric dimethylarginine) is a cardiovascular risk factor and an endogenous inhibitor of NOS (nitric oxide synthase). ADMA is metabolized by DDAHs (dimethylarginine dimethylaminohydrolases). ADMA levels are increased in cardiovascular disorders associated with abnormal angiogenesis but the mechanisms are poorly understood. Recent studies show that altering ADMA metabolism in vivo and in vitro modulates the activity of Rho GTPases, key regulators of actin dynamics, endothelial cell motility and angiogenesis. In the present review, we consider this and other NO-dependent and -independent molecular mechanisms by which the DDAH/ADMA pathway regulates angiogenesis.
Collapse
|
49
|
Leypoldt F, Choe CU, Gelderblom M, von Leitner EC, Atzler D, Schwedhelm E, Gerloff C, Sydow K, Böger RH, Magnus T. Dimethylarginine dimethylaminohydrolase-1 transgenic mice are not protected from ischemic stroke. PLoS One 2009; 4:e7337. [PMID: 19809508 PMCID: PMC2753663 DOI: 10.1371/journal.pone.0007337] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 09/11/2009] [Indexed: 11/19/2022] Open
Abstract
Background Methylated arginines are endogenous analogues of L-arginine, the substrate for nitric oxide (NO) synthase. Asymmetric dimethylarginine (ADMA) interferes with NO formation, causing endothelial dysfunction. ADMA is a predictor of cardiovascular events and mortality in humans. It is eliminated primarily by enzymatic activity of dimethylarginine dimethylaminohydrolase (DDAH). Methodology/Principal Findings We investigated whether human DDAH-1 (hDDAH-1) transgenicity protects from ischemic tissue damage in temporary middle cerebral artery occlusion (tMCAO) in mice. Infarct sizes did not significantly differ between hDDAH-1 transgenic (TG) mice and wild-type littermates (WT). As expected, ADMA plasma concentrations were significantly decreased, cerebral hDDAH expression and protein significantly increased in transgenic animals. Interestingly, neither brain tissue DDAH activity nor ADMA concentrations were different between TG and WT mice. In contrast, muscular DDAH activity was generally lower than in brain but significantly increased in TG mice. Conclusion/Significance Our study demonstrates that hDDAH-1 transgenic mice are not protected from ischemic cerebral tissue damage in tMCAO. This lack of protection is due to high basal cerebral DDAH activity, which is not further increasable by transgenic overexpression of DDAH.
Collapse
Affiliation(s)
- Frank Leypoldt
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fiedler LR, Bachetti T, Leiper J, Zachary I, Chen L, Renné T, Wojciak-Stothard B. The ADMA/DDAH pathway regulates VEGF-mediated angiogenesis. Arterioscler Thromb Vasc Biol 2009; 29:2117-24. [PMID: 19778944 DOI: 10.1161/atvbaha.109.194035] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Asymmetrical dimethylarginine (ADMA) is a nitric oxide synthase (NOS) inhibitor and cardiovascular risk factor associated with angiogenic disorders. Enzymes metabolising ADMA, dimethylarginine dimethylaminohydrolases (DDAH) promote angiogenesis, but the mechanisms are not clear. We hypothesized that ADMA/DDAH modifies endothelial responses to vascular endothelial growth factor (VEGF) by affecting activity of Rho GTPases, regulators of actin polymerization, and focal adhesion dynamics. METHODS AND RESULTS The effects of ADMA on VEGF-induced endothelial cell motility, focal adhesion turnover, and angiogenesis were studied in human umbilical vein endothelial cells (HUVECs) and DDAH I heterozygous knockout mice. ADMA inhibited VEGF-induced chemotaxis in vitro and angiogenesis in vitro and in vivo in an NO-dependent way. ADMA effects were prevented by overexpression of DDAH but were not associated with decreased proliferation, increased apoptosis, or changes in VEGFR-2 activity or expression. ADMA inhibited endothelial cell polarization, protrusion formation, and decreased focal adhesion dynamics, resulting from Rac1 inhibition after decrease in phosphorylation of vasodilator stimulated phosphoprotein (VASP). Constitutively active Rac1, and to a lesser extent dominant negative RhoA, abrogated ADMA effects in vitro and in vivo. CONCLUSIONS The ADMA/DDAH pathway regulates VEGF-induced angiogenesis in an NO- and Rac1-dependent manner.
Collapse
Affiliation(s)
- Lorna R Fiedler
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, UK
| | | | | | | | | | | | | |
Collapse
|