1
|
Heuser SK, Li J, Pudewell S, LoBue A, Li Z, Cortese-Krott MM. Biochemistry, pharmacology and in vivo function of arginases. Pharmacol Rev 2024; 77:PHARMREV-AR-2024-001271. [PMID: 39406506 DOI: 10.1124/pharmrev.124.001271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 01/22/2025] Open
Abstract
Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea. The two existing isoforms Arg1 and Arg2 show different cellular localizations and metabolic functions. Arginase activity is crucial for nitrogen detoxification in the urea cycle, synthesis of polyamines, and control of l-arginine bioavailability and nitric oxide production. Despite significant progress in the understanding of the biochemistry and function of arginases, several open questions remain. Recent studies have revealed that the regulation and function of Arg1 and Arg2 are cell-type-specific, species-specific, and profoundly different in mice and humans. The main differences were found in the distribution and function of Arg1 and Arg2 in immune and erythroid cells. Contrary to what was previously thought, Arg1 activity appears to be only partially related to vascular NO signaling under homeostatic conditions in the vascular wall, but its expression is increased under disease conditions and may be targeted by treatment with arginase inhibitors. Arg2 appears to be mainly a catabolic enzyme involved in the synthesis of L-ornithine, polyamine, and proline but may play a putative role in blood pressure control, at least in mice. The immunosuppressive role of arginase-mediated arginine depletion is a promising target for cancer treatment. This review critically revises and discusses the biochemistry, pharmacology, and in vivo function of arginase, focusing on the insights gained from the analysis of cell-specific Arg1 and Arg2 knockout mice and human studies using arginase inhibitors or pegylated recombinant arginase. Significance Statement The review emphasizes the need for further research to deepen our understanding of the regulation of Arg1 and Arg 2 in different cell types under consideration of their localization, species-specificity, and multiple biochemical and physiological roles. This could lead to better pharmacological strategies to target arginase activity in liver, cardiovascular, hematological, immune/infection diseases and cancer.
Collapse
Affiliation(s)
- Sophia K Heuser
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Junjie Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Silke Pudewell
- Department of Biochemistry and Molecular Biology II, University of Duesseldorf, Germany
| | - Anthea LoBue
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Zhixin Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| |
Collapse
|
2
|
Pandian K, Huang L, Junaid A, Harms A, van Zonneveld AJ, Hankemeier T. Tracer-based metabolomics for profiling nitric oxide metabolites in a 3D microvessels-on-chip model. FASEB J 2024; 38:e70005. [PMID: 39171967 DOI: 10.1096/fj.202400553r] [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: 03/12/2024] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Endothelial dysfunction, prevalent in cardiovascular diseases (CVDs) and linked to conditions like diabetes, hypertension, obesity, renal failure, or hypercholesterolemia, is characterized by diminished nitric oxide (NO) bioavailability-a key signaling molecule for vascular homeostasis. Current two-dimensional (2D) in vitro studies on NO synthesis by endothelial cells (ECs) lack the crucial laminar shear stress, a vital factor in modulating the NO-generating enzyme, endothelial nitric oxide synthase (eNOS), under physiological conditions. Here we developed a tracer-based metabolomics approach to measure NO-specific metabolites with mass spectrometry (MS) and show the impact of fluid flow on metabolic parameters associated with NO synthesis using 2D and 3D platforms. Specifically, we tracked the conversion of stable-isotope labeled NO substrate L-Arginine to L-Citrulline and L-Ornithine to determine eNOS activity. We demonstrated clear responses in human coronary artery endothelial cells (HCAECs) cultured with 13C6, 15N4-L-Arginine, and treated with eNOS stimulator, eNOS inhibitor, and arginase inhibitor. Analysis of downstream metabolites, 13C6, 15N3 L-Citrulline and 13C5, 15N2 L-Ornithine, revealed distinct outcomes. Additionally, we evaluated the NO metabolic status in static 2D culture and 3D microvessel models with bidirectional and unidirectional fluid flow. Our 3D model exhibited significant effects, particularly in microvessels exposed to the eNOS stimulator, as indicated by the 13C6, 15N3 L-Citrulline/13C5, 15N2 L-Ornithine ratio, compared to the 2D culture. The obtained results indicate that the 2D static culture mimics an endothelial dysfunction status, while the 3D model with a unidirectional fluid flow provides a more representative physiological environment that provides a better model to study endothelial dysfunction.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Luojiao Huang
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| |
Collapse
|
3
|
Merkhan M, Mohammad J, Fathi Z, Younis Z, Mahmood SM, Mohammed M. Silent hyperlipidaemia modulated vascular endothelial markers. PHARMACIA 2021. [DOI: 10.3897/pharmacia.68.e67959] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The aetiology of ischemic heart diseases is mainly based on atherosclerosis of coronary artery. Inflammation and oxidative reactions are initiating and aggravating the illness resulting in pathological remodelling of vasculaturze at site of injury. Endothelium lining of blood vessels participated in the reaction biochemically through releasing some proteins into circulatory system which further complicate the condition. The aim of this study was to determine early diagnosed hyperlipidaemia-associated changes of the plasma level of some of these endothelial biomolecules. Compared to healthy control, hyperlipidaemic patients have significantly increased arginase, metalloendopeptidase, peroxidase, myeloperoxidase, and peroxynitrite with concomitant reduction in arylesterase and nitric oxide. The present study concluded that hyperlipidaemia play a great role in modulation of certain plasma protein markers which might be directly related to patient pathological condition or could be used as a tool for diagnosis or patient follow up indicating the stage of vasculature remodelling, healing, inflammation or resolution.
Collapse
|
4
|
Michell DL, Shihata WA, Andrews KL, Abidin NAZ, Jefferis AM, Sampson AK, Lumsden NG, Huet O, Parat MO, Jennings GL, Parton RG, Woollard KJ, Kaye DM, Chin-Dusting JPF, Murphy AJ. High intraluminal pressure promotes vascular inflammation via caveolin-1. Sci Rep 2021; 11:5894. [PMID: 33723357 PMCID: PMC7960707 DOI: 10.1038/s41598-021-85476-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
The aetiology and progression of hypertension involves various endogenous systems, such as the renin angiotensin system, the sympathetic nervous system, and endothelial dysfunction. Recent data suggest that vascular inflammation may also play a key role in the pathogenesis of hypertension. This study sought to determine whether high intraluminal pressure results in vascular inflammation. Leukocyte adhesion was assessed in rat carotid arteries exposed to 1 h of high intraluminal pressure. The effect of intraluminal pressure on signaling mechanisms including reactive oxygen species production (ROS), arginase expression, and NFĸB translocation was monitored. 1 h exposure to high intraluminal pressure (120 mmHg) resulted in increased leukocyte adhesion and inflammatory gene expression in rat carotid arteries. High intraluminal pressure also resulted in a downstream signaling cascade of ROS production, arginase expression, and NFĸB translocation. This process was found to be angiotensin II-independent and mediated by the mechanosensor caveolae, as caveolin-1 (Cav1)-deficient endothelial cells and mice were protected from pressure-induced vascular inflammatory signaling and leukocyte adhesion. Cav1 deficiency also resulted in a reduction in pressure-induced glomerular macrophage infiltration in vivo. These findings demonstrate Cav1 is an important mechanosensor in pressure-induced vascular and renal inflammation.
Collapse
Affiliation(s)
- Danielle L Michell
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Waled A Shihata
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Medicine, Monash University, Clayton, VIC, Australia.
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia.
| | - Karen L Andrews
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Nurul Aisha Zainal Abidin
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | | | | | | | - Olivier Huet
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Marie-Odile Parat
- School of Pharmacy, University of Queensland, St Lucia, QLD, Australia
| | | | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland, St Lucia, QLD, Australia
| | - Kevin J Woollard
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - David M Kaye
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jaye P F Chin-Dusting
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Clayton, VIC, Australia
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
5
|
From Mitochondria to Atherosclerosis: The Inflammation Path. Biomedicines 2021; 9:biomedicines9030258. [PMID: 33807807 PMCID: PMC8000234 DOI: 10.3390/biomedicines9030258] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key process in metazoan organisms due to its relevance for innate defense against infections and tissue damage. However, inflammation is also implicated in pathological processes such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease of the arterial wall where unstable atherosclerotic plaque rupture causing platelet aggregation and thrombosis may compromise the arterial lumen, leading to acute or chronic ischemic syndromes. In this review, we will focus on the role of mitochondria in atherosclerosis while keeping inflammation as a link. Mitochondria are the main source of cellular energy. Under stress, mitochondria are also capable of controlling inflammation through the production of reactive oxygen species (ROS) and the release of mitochondrial components, such as mitochondrial DNA (mtDNA), into the cytoplasm or into the extracellular matrix, where they act as danger signals when recognized by innate immune receptors. Primary or secondary mitochondrial dysfunctions are associated with the initiation and progression of atherosclerosis by elevating the production of ROS, altering mitochondrial dynamics and energy supply, as well as promoting inflammation. Knowing and understanding the pathways behind mitochondrial-based inflammation in atheroma progression is essential to discovering alternative or complementary treatments.
Collapse
|
6
|
Acacetin improves endothelial dysfunction and aortic fibrosis in insulin-resistant SHR rats by estrogen receptors. Mol Biol Rep 2020; 47:6899-6918. [PMID: 32892299 PMCID: PMC7561596 DOI: 10.1007/s11033-020-05746-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/28/2020] [Indexed: 12/20/2022]
Abstract
The aim of the work was to investigate the effects of acacetin on endothelial dysfunction and aortic fibrosis in insulin-resistant SHR rats and explore its mechanism. Seven-week-old male spontaneously hypertensive rats (SHR) were selected to establish a rat model of hypertension with insulin resistance induced by 10% fructose. The nuclear factor kappa B p65 (NF-κB p65) and Collagen I were observed by Immunohistochemistry. Immunofluorescence was used to observe estrogen receptor-alpha (ERα), estrogen receptor-beta (ERβ), and G protein-coupled receptor 30 (GPR30). Western blotting was used to detect interleukin (IL-1β), Arginase 2 (ARG2), Nostrin, endothelial nitric oxide synthase (eNOS), TGF-β, Smad3, ERK pathway proteins such as p-c-Raf, p-MEK1/2, p-ERK, ERK, p-P90RSK and p-MSK1. We found that acacetin did have an improvement on endothelial dysfunction and fibrosis. Meanwhile, it was also found to have a significant effect on the level of estrogen in this model by accident. Then, the experiment of uterine weight gain in mice confirmed that acacetin had a certain estrogen-like effect in vivo and played its role through the estrogen receptors pathway. In vitro experience HUVEC cells were stimulated with 30 mM/L glucose and 100 mM/L NaCl for 24 h to establish the endothelial cell injury model. HUVEC cells were treated with 1 μM/L estrogen receptors antagonist (ICI 182780) for 30 min before administration. Cell experiments showed that acacetin could reduce the apoptosis of HUVEC cells, the levels of inflammatory cytokines and the expression of TGF-β, Collagen I and Smad3 in endothelial cell injury model. After treatment with ICI 182780, the improvement of acacetin was significantly reversed. The results showed that acacetin relieved endothelial dysfunction and reduced the aortic fibrosis in insulin-resistant SHR rats by reducing the release of inflammatory factors and improving vasodilatory function through estrogen signaling pathway.
Collapse
|
7
|
Luo L, Wang Y, Hu P, Wu J. Long Non-Coding RNA Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) Promotes Hypertension by Modulating the Hsa-miR-124-3p/Nuclear Receptor Subfamily 3, Group C, Member 2 (NR3C2) and Hsa-miR-135a-5p/NR3C2 Axis. Med Sci Monit 2020; 26:e920478. [PMID: 32222724 PMCID: PMC7139186 DOI: 10.12659/msm.920478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background This study was designed to investigate the role of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in the proliferation as well as apoptosis of human umbilical vein endothelial cells (HUVECs), to offer a basis for therapy of hypertension. Material/Methods The lncRNA MALAT1 expression, hsa-miR-124-3p, hsa-miR-135a-5p, hsa-miR-135b-5p, and hsa-miR-455-5p in plasma were measured from 230 patients with hypertension and 230 non-hypertensive controls. The mechanism for lncRNA MALAT1 modulating the proliferation and apoptosis of HUVECs was explored by cell transfection, Cell Counting Kit-8 (CCK-8), quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and dual-luciferase reporter assays. Results The expression of hsa-miR-124-3p and hsa-miR-135a-5p was reduced and the expression of lncRNA MALAT1 was increased in the plasma of hypertensive patients. Moreover, the plasma levels of hsa-miR-124-3p and hsa-miR-135a-5p of hypertensive patients were negatively correlated with lncRNA MALAT1 (r=−0.64, −0.72; P<0.01, P<0.01, respectively). The level of nuclear receptor subfamily 3, group C, member 2 (NR3C2) protein was negatively correlated with hsa-miR-124-3p and hsa-miR-135a-5p (r=−0.74, −0.84; P<0.01, P<0.01, respectively). The proliferation of HUVECs was inhibited after the inhibition of MALAT. Additionally, after knocking down MALAT, the levels of hsa-miR-124-3p and hsa-miR-135a-5p in HUVECs were markedly increased, while the expression level of NR3C2 protein was decreased. The apoptotic rate of HUVECs after the transfection of MALAT1 small interfering RNA (si-MALAT1) (3.64±0.21%) was significantly reduced compared to that of transfected si-MALAT1 no template control (NC) (3.76±0.19%) and the control group (10.51±1.24%). Conclusions LncRNA MALAT1 regulates proliferation and apoptosis of HUVECs through the hsa-miR-124-3p/NR3C2 and/or hsa-miR-135a-5p/NR3C2 axis.
Collapse
Affiliation(s)
- Liju Luo
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Yu Wang
- Department of Cardiology, The Affiliated Yueqing Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Pengfei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Jiale Wu
- Department of Geratology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
8
|
Durante W. Amino Acids in Circulatory Function and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:39-56. [PMID: 32761569 DOI: 10.1007/978-3-030-45328-2_3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is the major cause of global mortality and disability. Abundant evidence indicates that amino acids play a fundamental role in cardiovascular physiology and pathology. Decades of research established the importance of L-arginine in promoting vascular health through the generation of the gas nitric oxide. More recently, L-glutamine, L-tryptophan, and L-cysteine have also been shown to modulate vascular function via the formation of a myriad of metabolites, including a number of gases (ammonia, carbon monoxide, hydrogen sulfide, and sulfur dioxide). These amino acids and their metabolites preserve vascular homeostasis by regulating critical cellular processes including proliferation, migration, differentiation, apoptosis, contractility, and senescence. Furthermore, they exert potent anti-inflammatory and antioxidant effects in the circulation, and block the accumulation of lipids within the arterial wall. They also mitigate known risk factors for cardiovascular disease, including hypertension, hyperlipidemia, obesity, and diabetes. However, in some instances, the metabolism of these amino acids through discrete pathways yields compounds that fosters vascular disease. While supplementation with amino acid monotherapy targeting the deficiency has ameliorated arterial disease in many animal models, this approach has been less successful in the clinic. A more robust approach combining amino acid supplementation with antioxidants, anti-inflammatory agents, and/or specific amino acid enzymatic pathway inhibitors may prove more successful. Alternatively, supplementation with amino acid-derived metabolites rather than the parent molecule may elicit beneficial effects while bypassing potentially harmful pathways of metabolism. Finally, there is an emerging recognition that circulating levels of multiple amino acids are perturbed in vascular disease and that a more holistic approach that targets all these amino acid derangements is required to restore circulatory function in diseased blood vessels.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
9
|
Factors Enhancing Serum Syndecan-1 Concentrations: A Large-Scale Comprehensive Medical Examination. J Clin Med 2019; 8:jcm8091320. [PMID: 31462009 PMCID: PMC6780947 DOI: 10.3390/jcm8091320] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/20/2019] [Accepted: 08/24/2019] [Indexed: 12/22/2022] Open
Abstract
Endothelial disorders are related to various diseases. An initial endothelial injury is characterized by endothelial glycocalyx injury. We aimed to evaluate endothelial glycocalyx injury by measuring serum syndecan-1 concentrations in patients during comprehensive medical examinations. A single-center, prospective, observational study was conducted at Asahi University Hospital. The participants enrolled in this study were 1313 patients who underwent comprehensive medical examinations at Asahi University Hospital from January 2018 to June 2018. One patient undergoing hemodialysis was excluded from the study. At enrollment, blood samples were obtained, and study personnel collected demographic and clinical data. No treatments or exposures were conducted except for standard medical examinations and blood sample collection. Laboratory data were obtained by the collection of blood samples at the time of study enrolment. According to nonlinear regression, the concentrations of serum syndecan-1 were significantly related to age (p = 0.016), aspartic aminotransferase concentration (AST, p = 0.020), blood urea nitrogen concentration (BUN, p = 0.013), triglyceride concentration (p < 0.001), and hematocrit (p = 0.006). These relationships were independent associations. Endothelial glycocalyx injury, which is reflected by serum syndecan-1 concentrations, is related to age, hematocrit, AST concentration, BUN concentration, and triglyceride concentration.
Collapse
|
10
|
Peng W, Cai G, Xia Y, Chen J, Wu P, Wang Z, Li G, Wei D. Mitochondrial Dysfunction in Atherosclerosis. DNA Cell Biol 2019; 38:597-606. [PMID: 31095428 DOI: 10.1089/dna.2018.4552] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are highly dynamic organelles beyond powerhouses of a cell. These components also play important roles in cell homeostasis by regulating cell function and phenotypic modulation. Atherosclerosis is the leading cause of morbidity and mortality in developed and developing countries. Mitochondrial dysfunction has been increasingly associated with the initiation and progression of atherosclerosis by elevating the production of reactive oxygen species and mitochondrial oxidative stress damage, mitochondrial dynamics dysfunction, and energy supply. In this review, we describe the progression of the link between mitochondrial dysfunction and atherosclerosis and its potential regulation mechanisms.
Collapse
Affiliation(s)
- Wenxi Peng
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Guoding Cai
- 2 Department of Cardiothoracic Surgery, The First Affiliated Hospital of University of South China, Hengyang, P.R. China
| | - Yiping Xia
- 3 University of South China of Nursing, Hengyang, P.R. China
| | - Jinna Chen
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Peng Wu
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Zuo Wang
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Guohua Li
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Dangheng Wei
- 1 Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| |
Collapse
|
11
|
Tavares G, Venturini G, Padilha K, Zatz R, Pereira AC, Thadhani RI, Rhee EP, Titan SMO. 1,5-Anhydroglucitol predicts CKD progression in macroalbuminuric diabetic kidney disease: results from non-targeted metabolomics. Metabolomics 2018; 14:39. [PMID: 30830377 DOI: 10.1007/s11306-018-1337-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/06/2018] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Metabolomics allows exploration of novel biomarkers and provides insights on metabolic pathways associated with disease. To date, metabolomics studies on CKD have been largely limited to Caucasian populations and have mostly examined surrogate end points. OBJECTIVE In this study, we evaluated the role of metabolites in predicting a primary outcome defined as dialysis need, doubling of serum creatinine or death in Brazilian macroalbuminuric DKD patients. METHODS Non-targeted metabolomics was performed on plasma from 56 DKD patients. Technical triplicates were done. Metabolites were identified using Agilent Fiehn GC/MS Metabolomics and NIST libraries (Agilent MassHunter Work-station Quantitative Analysis, version B.06.00). After data cleaning, 186 metabolites were left for analyses. RESULTS During a median follow-up time of 2.5 years, the PO occurred in 17 patients (30.3%). In non-parametric testing, 13 metabolites were associated with the PO. In univariate Cox regression, only 1,5-anhydroglucitol (HR 0.10; 95% CI 0.01-0.63, p = .01), norvaline and L-aspartic acid were associated with the PO. After adjustment for baseline renal function, 1,5-anhydroglucitol (HR 0.10; 95% CI 0.02-0.63, p = .01), norvaline (HR 0.01; 95% CI 0.001-0.4, p = .01) and aspartic acid (HR 0.12; 95% CI 0.02-0.64, p = .01) remained significantly and inversely associated with the PO. CONCLUSION Our results show that lower levels of 1,5-anhydroglucitol, norvaline and L-aspartic acid are associated with progression of macroalbuminuric DKD. While norvaline and L-aspartic acid point to interesting metabolic pathways, 1,5-anhydroglucitol is of particular interest since it has been previously shown to be associated with incident CKD. This inverse biomarker of hyperglycemia should be further explored as a new tool in DKD.
Collapse
Affiliation(s)
- Gesiane Tavares
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil.
| | - Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Roberto Zatz
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Ravi I Thadhani
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eugene P Rhee
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Silvia M O Titan
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil
| |
Collapse
|
12
|
Xiong Y, Yepuri G, Necetin S, Montani JP, Ming XF, Yang Z. Arginase-II Promotes Tumor Necrosis Factor-α Release From Pancreatic Acinar Cells Causing β-Cell Apoptosis in Aging. Diabetes 2017; 66:1636-1649. [PMID: 28356309 DOI: 10.2337/db16-1190] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/21/2017] [Indexed: 11/13/2022]
Abstract
Aging is associated with glucose intolerance. Arginase-II (Arg-II), the type-II L-arginine-ureahydrolase, is highly expressed in pancreas. However, its role in regulation of pancreatic β-cell function is not known. Here we show that female (not male) mice deficient in Arg-II (Arg-II-/-) are protected from age-associated glucose intolerance and reveal greater glucose induced-insulin release, larger islet size and β-cell mass, and more proliferative and less apoptotic β-cells compared with the age-matched wild-type (WT) controls. Moreover, Arg-II is mainly expressed in acinar cells and is upregulated with aging, which enhances p38 mitogen-activated protein kinase (p38 MAPK) activation and release of tumor necrosis factor-α (TNF-α). Accordingly, conditioned medium of isolated acinar cells from old WT (not Arg-II-/-) mice contains higher TNF-α levels than the young mice and stimulates β-cell apoptosis and dysfunction, which are prevented by a neutralizing anti-TNF-α antibody. In acinar cells, our study demonstrates an age-associated Arg-II upregulation, which promotes TNF-α release through p38 MAPK leading to β-cell apoptosis, insufficient insulin secretion, and glucose intolerance in female rather than male mice.
Collapse
Affiliation(s)
- Yuyan Xiong
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
- Kidney Control of Homeostasis, Swiss National Centre of Competence in Research, Zurich, Switzerland
| | - Gautham Yepuri
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
| | - Sevil Necetin
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
| | - Jean-Pierre Montani
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
- Kidney Control of Homeostasis, Swiss National Centre of Competence in Research, Zurich, Switzerland
| | - Xiu-Fen Ming
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
- Kidney Control of Homeostasis, Swiss National Centre of Competence in Research, Zurich, Switzerland
| | - Zhihong Yang
- Cardiovascular and Aging Research, Department of Medicine, Division of Physiology, University of Fribourg, Fribourg, Switzerland
- Kidney Control of Homeostasis, Swiss National Centre of Competence in Research, Zurich, Switzerland
| |
Collapse
|
13
|
The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activator dh404 protects against diabetes-induced endothelial dysfunction. Cardiovasc Diabetol 2017; 16:33. [PMID: 28253885 PMCID: PMC5335831 DOI: 10.1186/s12933-017-0513-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/22/2017] [Indexed: 12/13/2022] Open
Abstract
Background Vascular dysfunction is a pivotal event in the development of diabetes-associated vascular disease. Increased inflammation and oxidative stress are major contributors to vascular dysfunction. Nrf2, a master regulator of several anti-oxidant genes and a suppressor of inflammatory NF-κB, has potential as a target to combat oxidative stress and inflammation. The aim of this study was to investigate the effects of a novel Nrf2 activator, the bardoxolone methyl derivative dh404, on endothelial function in vitro and in vivo. Methods dh404 at 3 mg/kg was administered to male Akita mice, an established diabetic mouse model of insulin insufficiency and hyperglycemia, from 6 weeks of age. At 26 weeks of age, vascular reactivity was assessed by wire myography, pro-inflammatory expression was assessed in the aortas by qRT-PCR and immunohistochemistry, and systemic and vascular oxidative stress measurements were determined. Additionally, studies in human aortic endothelial cells (HAECs) derived from normal and diabetic patients in the presence or absence of dh404 included assessment of pro-inflammatory genes by qRT-PCR and western blotting. Oxidative stress was assessed by three methods; L-012, DCFDA and amplex red. Static adhesion assays were performed to determine the leukocyte–endothelial interaction in the presence or absence of dh404. Results Dh404 significantly attenuated endothelial dysfunction in diabetic Akita mice characterized by reduced contraction in response to phenylephrine and the downregulation of inflammatory genes (VCAM-1, ICAM-1, p65, IL-1β) and pro-oxidant genes (Nox1 and Nox2). Furthermore, reduced systemic and vascular oxidative stress levels were observed in diabetic Akita mice. dh404 exhibited cytoprotective effects in diabetic HAECs in vitro, reflected by significant upregulation of Nrf2-responsive genes, NAD(P)H quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), reduction of oxidative stress markers (O2·− and H2O2), inhibition of inflammatory genes (VCAM-1 and the p65 subunit of NF-κB) and attenuation of leukocyte–endothelial interactions (P < 0.05 for all in vitro and in vivo parameters; one or two-way ANOVA as appropriate with post hoc testing). Conclusion These studies demonstrate that upregulation of Nrf2 by dh404 represents a novel therapeutic strategy to limit diabetes-associated vascular injury. Electronic supplementary material The online version of this article (doi:10.1186/s12933-017-0513-y) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ Res 2017; 120:713-735. [DOI: 10.1161/circresaha.116.309326] [Citation(s) in RCA: 692] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
Major reactive oxygen species (ROS)–producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).
Collapse
Affiliation(s)
- Ulrich Förstermann
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Ning Xia
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Huige Li
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| |
Collapse
|
15
|
Chung BL, Toth MJ, Kamaly N, Sei YJ, Becraft J, Mulder WJM, Fayad ZA, Farokhzad OC, Kim Y, Langer R. Nanomedicines for Endothelial Disorders. NANO TODAY 2015; 10:759-776. [PMID: 26955397 PMCID: PMC4778260 DOI: 10.1016/j.nantod.2015.11.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The endothelium lines the internal surfaces of blood and lymphatic vessels and has a critical role in maintaining homeostasis. Endothelial dysfunction is involved in the pathology of many diseases and conditions, including disorders such as diabetes, cardiovascular diseases, and cancer. Given this common etiology in a range of diseases, medicines targeting an impaired endothelium can strengthen the arsenal of therapeutics. Nanomedicine - the application of nanotechnology to healthcare - presents novel opportunities and potential for the treatment of diseases associated with an impaired endothelium. This review discusses therapies currently available for the treatment of these disorders and highlights the application of nanomedicine for the therapy of these major disease complications.
Collapse
Affiliation(s)
- Bomy Lee Chung
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
- Department of Chemical Engineering, Massachusetts Institute of Technology
| | - Michael J. Toth
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Institute for Electronics and Nanotechnology (IEN), Parker H. Petit Institute for Bioengineering and Bioscience (IBB), Georgia Institute of Technology
| | - Nazila Kamaly
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
- Laboratory of Nanomedicine and Biomaterials, Brigham and Women’s Hospital, Harvard Medical School
| | - Yoshitaka J. Sei
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Institute for Electronics and Nanotechnology (IEN), Parker H. Petit Institute for Bioengineering and Bioscience (IBB), Georgia Institute of Technology
| | - Jacob Becraft
- Department of Biological Engineering, Massachusetts Institute of Technology
| | - Willem J. M. Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai
| | - Zahi A. Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai
| | - Omid C. Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Brigham and Women’s Hospital, Harvard Medical School
- King Abdulaziz University, Jeddah, Saudi Arabia
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, Institute for Electronics and Nanotechnology (IEN), Parker H. Petit Institute for Bioengineering and Bioscience (IBB), Georgia Institute of Technology
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology
- Department of Chemical Engineering, Massachusetts Institute of Technology
- Department of Biological Engineering, Massachusetts Institute of Technology
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology
| |
Collapse
|
16
|
Astigiano S, Morini M, Damonte P, Fraternali Orcioni G, Cassanello M, Puglisi A, Noonan DM, Bronte V, Barbieri O. Transgenic mice overexpressing arginase 1 in monocytic cell lineage are affected by lympho-myeloproliferative disorders and disseminated intravascular coagulation. Carcinogenesis 2015; 36:1354-62. [PMID: 26363032 DOI: 10.1093/carcin/bgv129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 08/27/2015] [Indexed: 11/12/2022] Open
Abstract
Arginase (ARG) is a metabolic enzyme present in two isoforms that hydrolyze l-arginine to urea and ornithine. In humans, ARG isoform 1 is also expressed in cells of the myeloid lineage. ARG activity promotes tumour growth and inhibits T lymphocyte activation. However, the two ARG transgenic mouse lines produced so far failed to show such effects. We have generated, in two different genetic backgrounds, transgenic mice constitutively expressing ARG1 under the control of the CD68 promoter in macrophages and monocytes. Both heterozygous and homozygous transgenic mice showed a relevant increase in mortality at early age, compared with wild-type siblings (67/267 and 48/181 versus 8/149, respectively, both P < 0.005). This increase was due to high incidence of haematologic malignancies, in particular myeloid leukaemia, myeloid dysplasia, lymphomas and disseminated intravascular coagulation (DIC), diseases that were absent in wild-type mice. Atrophy of lymphoid organs due to reduction in T-cell compartment was also detected. Our results indicate that ARG activity may participate in the pathogenesis of lymphoproliferative and myeloproliferative disorders, suggest the involvement of alterations of L-arginine metabolism in the onset of DIC and confirm a role for the enzyme in regulating T-cell homeostasis.
Collapse
Affiliation(s)
- Simonetta Astigiano
- Department of Haematology and Oncology, IRCCS AOU San Martino-IST National Institute for Cancer Research, Genova 16132, Italy
| | - Monica Morini
- Department of Experimental Medicine, University of Genova, Genova 16132, Italy, Present address: IIT-The Italian Institute of Technology, Genova, Italy
| | - Patrizia Damonte
- Department of Experimental Medicine, University of Genova, Genova 16132, Italy
| | - Giulio Fraternali Orcioni
- Department of Haematology and Oncology, IRCCS AOU San Martino-IST National Institute for Cancer Research, Genova 16132, Italy
| | - Michela Cassanello
- Laboratory for the Study of Inborn Errors of Metabolism, Istituto Giannina Gaslini, Genova 16100, Italy
| | - Andrea Puglisi
- Department of Experimental Medicine, University of Genova, Genova 16132, Italy
| | - Douglas M Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese 21100, Italy, Department of Oncology, IRCCS MultiMedica, 20099 Sesto San Giovanni, Milan, Italy and
| | - Vincenzo Bronte
- Department of Pathology and Diagnostic, University Hospital, Verona 37100, Italy
| | - Ottavia Barbieri
- Department of Haematology and Oncology, IRCCS AOU San Martino-IST National Institute for Cancer Research, Genova 16132, Italy, Department of Experimental Medicine, University of Genova, Genova 16132, Italy,
| |
Collapse
|
17
|
Arginase as a Critical Prooxidant Mediator in the Binomial Endothelial Dysfunction-Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:924860. [PMID: 26064427 PMCID: PMC4434223 DOI: 10.1155/2015/924860] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/11/2014] [Indexed: 12/31/2022]
Abstract
Arginase is a metalloenzyme which hydrolyzes L-arginine to L-ornithine and urea. Since its discovery, in the early 1900s, this enzyme has gained increasing attention, as literature reports have progressively pointed to its critical participation in regulating nitric oxide bioavailability. Indeed, accumulating evidence in the following years would picture arginase as a key player in vascular health. Recent studies have highlighted the arginase regulatory role in the progression of atherosclerosis, the latter an essentially prooxidant state. Apart from the fact that arginase has been proven to impair different metabolic pathways, and also as a consequence of this, the repercussions of the actions of such enzyme go further than first thought. In fact, such metalloenzyme exhibits direct implications in multiple cardiometabolic diseases, among which are hypertension, type 2 diabetes, and hypercholesterolemia. Considering the epidemiological repercussions of these clinical conditions, arginase is currently seen under the spotlights of the search for developing specific inhibitors, in order to mitigate its deleterious effects. That said, the present review focuses on the role of arginase in endothelial function and its participation in the establishment of atherosclerotic lesions, discussing the main regulatory mechanisms of the enzyme, also highlighting the potential development of pharmacological strategies in related cardiovascular diseases.
Collapse
|
18
|
Ashmore T, Fernandez BO, Branco-Price C, West JA, Cowburn AS, Heather LC, Griffin JL, Johnson RS, Feelisch M, Murray AJ. Dietary nitrate increases arginine availability and protects mitochondrial complex I and energetics in the hypoxic rat heart. J Physiol 2014; 592:4715-31. [PMID: 25172947 PMCID: PMC4253472 DOI: 10.1113/jphysiol.2014.275263] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 08/18/2014] [Indexed: 12/31/2022] Open
Abstract
Hypoxic exposure is associated with impaired cardiac energetics in humans and altered mitochondrial function, with suppressed complex I-supported respiration, in rat heart. This response might limit reactive oxygen species generation, but at the cost of impaired electron transport chain (ETC) activity. Dietary nitrate supplementation improves mitochondrial efficiency and can promote tissue oxygenation by enhancing blood flow. We therefore hypothesised that ETC dysfunction, impaired energetics and oxidative damage in the hearts of rats exposed to chronic hypoxia could be alleviated by sustained administration of a moderate dose of dietary nitrate. Male Wistar rats (n = 40) were given water supplemented with 0.7 mmol l(-1) NaCl (as control) or 0.7 mmol l(-1) NaNO3, elevating plasma nitrate levels by 80%, and were exposed to 13% O2 (hypoxia) or normoxia (n = 10 per group) for 14 days. Respiration rates, ETC protein levels, mitochondrial density, ATP content and protein carbonylation were measured in cardiac muscle. Complex I respiration rates and protein levels were 33% lower in hypoxic/NaCl rats compared with normoxic/NaCl controls. Protein carbonylation was 65% higher in hearts of hypoxic rats compared with controls, indicating increased oxidative stress, whilst ATP levels were 62% lower. Respiration rates, complex I protein and activity, protein carbonylation and ATP levels were all fully protected in the hearts of nitrate-supplemented hypoxic rats. Both in normoxia and hypoxia, dietary nitrate suppressed cardiac arginase expression and activity and markedly elevated cardiac l-arginine concentrations, unmasking a novel mechanism of action by which nitrate enhances tissue NO bioavailability. Dietary nitrate therefore alleviates metabolic abnormalities in the hypoxic heart, improving myocardial energetics.
Collapse
Affiliation(s)
- Tom Ashmore
- Department of Physiology, Development & Neuroscience, University of Cambridge, UK Department of Biochemistry, University of Cambridge, UK
| | | | | | - James A West
- Department of Biochemistry, University of Cambridge, UK MRC-Human Nutrition Research, Elsie Widdowson Laboratory, Cambridge, UK
| | - Andrew S Cowburn
- Department of Physiology, Development & Neuroscience, University of Cambridge, UK
| | - Lisa C Heather
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, UK MRC-Human Nutrition Research, Elsie Widdowson Laboratory, Cambridge, UK
| | - Randall S Johnson
- Department of Physiology, Development & Neuroscience, University of Cambridge, UK
| | - Martin Feelisch
- Faculty of Medicine, Clinical & Experimental Sciences, University of Southampton, UK
| | - Andrew J Murray
- Department of Physiology, Development & Neuroscience, University of Cambridge, UK
| |
Collapse
|
19
|
Yang Z, Ming XF. Functions of arginase isoforms in macrophage inflammatory responses: impact on cardiovascular diseases and metabolic disorders. Front Immunol 2014; 5:533. [PMID: 25386179 PMCID: PMC4209887 DOI: 10.3389/fimmu.2014.00533] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/09/2014] [Indexed: 12/18/2022] Open
Abstract
Macrophages play a paramount role in immunity and inflammation-associated diseases, including infections, cardiovascular diseases, obesity-associated metabolic imbalances, and cancer. Compelling evidence from studies of recent years demonstrates that macrophages are heterogeneous and undergo heterogeneous phenotypic changes in response to microenvironmental stimuli. The M1 killer type response and the M2 repair type response are best known, and are two extreme examples. Among other markers, inducible nitric oxide synthase and type-I arginase (Arg-I), the enzymes that are involved in l-arginine/nitric oxide (NO) metabolism, are associated with the M1 and M2 phenotype, respectively, and therefore widely used as the markers for characterization of the two macrophage phenotypes. There is also a type-II arginase (Arg-II), which is expressed in macrophages and prevalently viewed as having the same function as Arg-I in the cells. In contrast to Arg-I, little information on the role of Arg-II in macrophage inflammatory responses is available. Emerging evidence, however, suggests that differential roles of Arg-I and Arg-II in regulating macrophage functions. In this article, we will review recent developments on the functional roles of the two arginase isoforms in regulation of macrophage inflammatory responses by focusing on their impact on the pathogenesis of cardiovascular diseases and metabolic disorders.
Collapse
Affiliation(s)
- Zhihong Yang
- Vascular Biology, Division of Physiology, Department of Medicine, Faculty of Science, University of Fribourg , Fribourg , Switzerland
| | - Xiu-Fen Ming
- Vascular Biology, Division of Physiology, Department of Medicine, Faculty of Science, University of Fribourg , Fribourg , Switzerland
| |
Collapse
|
20
|
Pandey D, Bhunia A, Oh YJ, Chang F, Bergman Y, Kim JH, Serbo J, Boronina TN, Cole RN, Van Eyk J, Remaley AT, Berkowitz DE, Romer LH. OxLDL triggers retrograde translocation of arginase2 in aortic endothelial cells via ROCK and mitochondrial processing peptidase. Circ Res 2014; 115:450-9. [PMID: 24903103 PMCID: PMC8760889 DOI: 10.1161/circresaha.115.304262] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Increased arginase activity contributes to endothelial dysfunction by competition for l-arginine substrate and reciprocal regulation of nitric oxide synthase (NOS). The rapid increase in arginase activity in human aortic endothelial cells exposed to oxidized low-density lipoprotein (OxLDL) is consistent with post-translational modification or subcellular trafficking. OBJECTIVE To test the hypotheses that OxLDL triggers reverse translocation of mitochondrial arginase 2 (Arg2) to cytosol and Arg2 activation, and that this process is dependent on mitochondrial processing peptidase, lectin-like OxLDL receptor-1 receptor, and rho kinase. METHODS AND RESULTS OxLDL-triggered translocation of Arg2 from mitochondria to cytosol in human aortic endothelial cells and in murine aortic intima with a concomitant rise in arginase activity. All of these changes were abolished by inhibition of mitochondrial processing peptidase or by its siRNA-mediated knockdown. Rho kinase inhibition and the absence of the lectin-like OxLDL receptor-1 in knockout mice also ablated translocation. Aminoterminal sequencing of Arg2 revealed 2 candidate mitochondrial targeting sequences, and deletion of either of these confined Arg2 to the cytoplasm. Inhibitors of mitochondrial processing peptidase or lectin-like OxLDL receptor-1 knockout attenuated OxLDL-mediated decrements in endothelial-specific NO production and increases in superoxide generation. Finally, Arg2(-/-) mice bred on an ApoE(-/-) background showed reduced plaque load, reduced reactive oxygen species production, enhanced NO, and improved endothelial function when compared with ApoE(-/-) controls. CONCLUSIONS These data demonstrate dual distribution of Arg2, a protein with an unambiguous mitochondrial targeting sequence, in mammalian cells, and its reverse translocation to cytoplasm by alterations in the extracellular milieu. This novel molecular mechanism drives OxLDL-mediated arginase activation, endothelial NOS uncoupling, endothelial dysfunction, and atherogenesis.
Collapse
Affiliation(s)
- Deepesh Pandey
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Anil Bhunia
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Young Jun Oh
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Fumin Chang
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Yehudit Bergman
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Jae Hyung Kim
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Janna Serbo
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Tatiana N Boronina
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Robert N Cole
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Jennifer Van Eyk
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Alan T Remaley
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Dan E Berkowitz
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.)
| | - Lewis H Romer
- From the Department of Anesthesiology and Critical Care Medicine (D.P., A.B., Y.J.O., F.C., Y.B., J.H.K., J.S., D.E.B., L.H.R.), Biomedical Engineering (J.S., D.E.B., L.H.R.), and Cell Biology, Pediatrics, Center for Cell Dynamics (L.H.R.), Mass Spectrometry and Proteomics Facility (T.N.B., R.N.C.), and Departments of Medicine and Biological Chemistry (J.V.E.), Johns Hopkins University School of Medicine, Baltimore, MD; and Lipoprotein Metabolism Section, Cardiovascular-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD (A.T.R.).
| |
Collapse
|
21
|
Tang X, Luo YX, Chen HZ, Liu DP. Mitochondria, endothelial cell function, and vascular diseases. Front Physiol 2014; 5:175. [PMID: 24834056 PMCID: PMC4018556 DOI: 10.3389/fphys.2014.00175] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/16/2014] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are perhaps the most sophisticated and dynamic responsive sensing systems in eukaryotic cells. The role of mitochondria goes beyond their capacity to create molecular fuel and includes the generation of reactive oxygen species, the regulation of calcium, and the activation of cell death. In endothelial cells, mitochondria have a profound impact on cellular function under both healthy and diseased conditions. In this review, we summarize the basic functions of mitochondria in endothelial cells and discuss the roles of mitochondria in endothelial dysfunction and vascular diseases, including atherosclerosis, diabetic vascular dysfunction, pulmonary artery hypertension, and hypertension. Finally, the potential therapeutic strategies to improve mitochondrial function in endothelial cells and vascular diseases are also discussed, with a focus on mitochondrial-targeted antioxidants and calorie restriction.
Collapse
Affiliation(s)
- Xiaoqiang Tang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - Yu-Xuan Luo
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| |
Collapse
|
22
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
23
|
|
24
|
Kuo L, Hein TW. Vasomotor regulation of coronary microcirculation by oxidative stress: role of arginase. Front Immunol 2013; 4:237. [PMID: 23966996 PMCID: PMC3746455 DOI: 10.3389/fimmu.2013.00237] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/01/2013] [Indexed: 12/14/2022] Open
Abstract
Overproduction of reactive oxygen species, i.e., oxidative stress, is associated with the activation of redox signaling pathways linking to inflammatory insults and cardiovascular diseases by impairing endothelial function and consequently blood flow dysregulation due to microvascular dysfunction. This review focuses on the regulation of vasomotor function in the coronary microcirculation by endothelial nitric oxide (NO) during oxidative stress and inflammation related to the activation of L-arginine consuming enzyme arginase. Superoxide produced in the vascular wall compromises vasomotor function by not only scavenging endothelium-derived NO but also inhibiting prostacyclin synthesis due to formation of peroxynitrite. The upregulation of arginase contributes to the deficiency of endothelial NO and microvascular dysfunction in various vascular diseases by initiating or following oxidative stress and inflammation. Hydrogen peroxide, a diffusible and stable oxidizing agent, exerts vasodilator function and plays important roles in the physiological regulation of coronary blood flow. In occlusive coronary ischemia, the release of hydrogen peroxide from the microvasculature helps to restore vasomotor function of coronary collateral microvessels with exercise training. However, excessive production and prolonged exposure of microvessels to hydrogen peroxide impairs NO-mediated endothelial function by reducing L-arginine availability through hydroxyl radical-dependent upregulation of arginase. The redox signaling can be a double-edged sword in the microcirculation, which helps tissue survival in one way by improving vasomotor regulation and elicits oxidative stress and tissue injury in the other way by causing vascular dysfunction. The impact of vascular arginase on the development of vasomotor dysfunction associated with angiotensin II receptor activation, hypertension, ischemia-reperfusion, hypercholesterolemia, and inflammatory insults is discussed.
Collapse
Affiliation(s)
- Lih Kuo
- Department of Medical Physiology, Scott & White Healthcare, Texas A&M Health Science Center, Temple, TX, USA
- Department of Surgery, College of Medicine, Scott & White Healthcare, Texas A&M Health Science Center, Temple, TX, USA
| | - Travis W. Hein
- Department of Surgery, College of Medicine, Scott & White Healthcare, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
25
|
Yang Z, Ming XF. Arginase: the emerging therapeutic target for vascular oxidative stress and inflammation. Front Immunol 2013; 4:149. [PMID: 23781221 PMCID: PMC3679468 DOI: 10.3389/fimmu.2013.00149] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/30/2013] [Indexed: 01/28/2023] Open
Abstract
Oxidative stress and inflammation in the vascular wall are essential mechanisms of atherosclerosis and vascular dysfunctions associated with risk factors such as metabolic diseases, aging, hypertension, etc. Evidence has been provided that activation of the vascular endothelial cells in the presence of the risk factors promotes oxidative stress and vascular inflammatory responses, leading to acceleration of atherosclerotic vascular disease. Increasing number of studies from recent years demonstrates that uncoupling of endothelial nitric oxide synthase (eNOS), whereby the enzyme eNOS produces detrimental amount of superoxide anion O2− instead the vasoprotective nitric oxide (NO⋅), plays a critical role in vascular dysfunction under various pathophysiological conditions and in aging. The mechanisms of eNOS-uncoupling seem multiple and complex. Recent research provides emerging evidence supporting an essential role of increased activity of arginases including arginase-I and arginase-II in causing eNOS-uncoupling, which results in vascular oxidative stress and inflammatory responses, and ultimately leading to vascular diseases. This review article will summarize the most recent findings on the functional roles of arginases in vascular diseases and/or dysfunctions and the underlying mechanisms in relation to oxidative stress and inflammations. Moreover, regulatory mechanisms of arginases in the vasculature are reviewed and the future perspectives of targeting arginases as therapeutic options in vascular diseases are discussed.
Collapse
Affiliation(s)
- Zhihong Yang
- Vascular Biology, Division of Physiology, Department of Medicine, University of Fribourg , Fribourg , Switzerland
| | | |
Collapse
|
26
|
Abstract
Arginase metabolizes the semi-essential amino acid l-arginine to l-ornithine and urea. There are two distinct isoforms of arginase, arginase I and II, which are encoded by separate genes and display differences in tissue distribution, subcellular localization, and molecular regulation. Blood vessels express both arginase I and II but their distribution appears to be cell-, vessel-, and species-specific. Both isoforms of arginase are induced by numerous pathologic stimuli and contribute to vascular cell dysfunction and vessel wall remodeling in several diseases. Clinical and experimental studies have documented increases in the expression and/or activity of arginase I or II in blood vessels following arterial injury and in pulmonary and arterial hypertension, aging, and atherosclerosis. Significantly, pharmacological inhibition or genetic ablation of arginase in animals ameliorates abnormalities in vascular cells and normalizes blood vessel architecture and function in all of these pathological states. The detrimental effect of arginase in vascular remodeling is attributable to its ability to stimulate vascular smooth muscle cell and endothelial cell proliferation, and collagen deposition by promoting the synthesis of polyamines and l-proline, respectively. In addition, arginase adversely impacts arterial remodeling by directing macrophages toward an inflammatory phenotype. Moreover, the proliferative, fibrotic, and inflammatory actions of arginase in the vasculature are further amplified by its capacity to inhibit nitric oxide (NO) synthesis by competing with NO synthase for substrate, l-arginine. Pharmacologic or molecular approaches targeting specific isoforms of arginase represent a promising strategy in treating obstructive fibroproliferative vascular disease.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia Columbia, MO, USA
| |
Collapse
|
27
|
Shemyakin A, Kövamees O, Rafnsson A, Böhm F, Svenarud P, Settergren M, Jung C, Pernow J. Arginase Inhibition Improves Endothelial Function in Patients With Coronary Artery Disease and Type 2 Diabetes Mellitus. Circulation 2012. [PMID: 23183942 DOI: 10.1161/circulationaha.112.140335] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background—
Endothelial dysfunction plays an important role in the early development of atherosclerosis and vascular complications in type 2 diabetes mellitus. Increased expression and activity of arginase, metabolizing the nitric oxide substrate
l
-arginine, may result in reduced production of nitric oxide and thereby endothelial dysfunction. We hypothesized that inhibition of arginase activity improves endothelial function in patients with coronary artery disease (CAD) and type 2 diabetes mellitus.
Methods and Results—
Three groups of subjects were included: 16 patients with CAD, 16 patients with CAD and type 2 diabetes mellitus (CAD+Diabetes), and 16 age-matched healthy control subjects. Forearm endothelium-dependent and endothelium-independent vasodilatation were assessed with venous occlusion plethysmography before and during intra-arterial infusion of the arginase inhibitor
N
ω
-hydroxy-nor-
l
-arginine (nor-NOHA; 0.1 mg/min). Nor-NOHA was also coinfused with the nitric oxide synthase inhibitor (
N
G
-monomethyl L-arginine). The expression of arginase was determined in the internal mammary artery of patients undergoing bypass surgery. Nor-NOHA markedly increased endothelium-dependent vasodilatation (up to 2-fold) in patients with CAD+Diabetes and CAD (
P
<0.001) but not in the control group.
N
G
-monomethyl L-arginine completely inhibited the increase in endothelium-dependent vasodilatation induced by nor-NOHA. Endothelium-independent vasodilatation was slightly improved by nor-NOHA in the CAD+Diabetes group. Arginase I was expressed in vascular smooth muscle cells and endothelial cells, and arginase II was expressed in endothelial cells of patients with and without diabetes mellitus.
Conclusions—
Arginase inhibition markedly improves endothelial function in patients with CAD and type 2 diabetes mellitus suggesting that increased arginase activity is a key factor in the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Alexey Shemyakin
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Oskar Kövamees
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Arnar Rafnsson
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Felix Böhm
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Peter Svenarud
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Magnus Settergren
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - Christian Jung
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| | - John Pernow
- From the Karolinska Institute, Department of Medicine, Division of Cardiology (A.S., O.K., A.R., F.B., M.S., C.J., J.P.), Department of Molecular Medicine and Surgery, Division of Thoracic Surgery (P.S.), Karolinska University Hospital, Stockholm, Sweden; and Department of Internal Medicine I, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (C.J.)
| |
Collapse
|