1
|
Martí I Líndez AA, Reith W. Arginine-dependent immune responses. Cell Mol Life Sci 2021; 78:5303-5324. [PMID: 34037806 PMCID: PMC8257534 DOI: 10.1007/s00018-021-03828-4] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
A growing body of evidence indicates that, over the course of evolution of the immune system, arginine has been selected as a node for the regulation of immune responses. An appropriate supply of arginine has long been associated with the improvement of immune responses. In addition to being a building block for protein synthesis, arginine serves as a substrate for distinct metabolic pathways that profoundly affect immune cell biology; especially macrophage, dendritic cell and T cell immunobiology. Arginine availability, synthesis, and catabolism are highly interrelated aspects of immune responses and their fine-tuning can dictate divergent pro-inflammatory or anti-inflammatory immune outcomes. Here, we review the organismal pathways of arginine metabolism in humans and rodents, as essential modulators of the availability of this semi-essential amino acid for immune cells. We subsequently review well-established and novel findings on the functional impact of arginine biosynthetic and catabolic pathways on the main immune cell lineages. Finally, as arginine has emerged as a molecule impacting on a plethora of immune functions, we integrate key notions on how the disruption or perversion of arginine metabolism is implicated in pathologies ranging from infectious diseases to autoimmunity and cancer.
Collapse
Affiliation(s)
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Redox and Antioxidant Modulation of Circadian Rhythms: Effects of Nitroxyl, N-Acetylcysteine and Glutathione. Molecules 2021; 26:molecules26092514. [PMID: 33925826 PMCID: PMC8123468 DOI: 10.3390/molecules26092514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/16/2022] Open
Abstract
The circadian clock at the hypothalamic suprachiasmatic nucleus (SCN) entrains output rhythms to 24-h light cycles. To entrain by phase-advances, light signaling at the end of subjective night (circadian time 18, CT18) requires free radical nitric oxide (NO•) binding to soluble guanylate cyclase (sGC) heme group, activating the cyclic guanosine monophosphate (cGMP)-dependent protein kinase (PKG). Phase-delays at CT14 seem to be independent of NO•, whose redox-related species were yet to be investigated. Here, the one-electron reduction of NO• nitroxyl was pharmacologically delivered by Angeli’s salt (AS) donor to assess its modulation on phase-resetting of locomotor rhythms in hamsters. Intracerebroventricular AS generated nitroxyl at the SCN, promoting phase-delays at CT14, but potentiated light-induced phase-advances at CT18. Glutathione/glutathione disulfide (GSH/GSSG) couple measured in SCN homogenates showed higher values at CT14 (i.e., more reduced) than at CT18 (oxidized). In addition, administration of antioxidants N-acetylcysteine (NAC) and GSH induced delays per se at CT14 but did not affect light-induced advances at CT18. Thus, the relative of NO• nitroxyl generates phase-delays in a reductive SCN environment, while an oxidative favors photic-advances. These data suggest that circadian phase-locking mechanisms should include redox SCN environment, generating relatives of NO•, as well as coupling with the molecular oscillator.
Collapse
|
3
|
Visible light-controlled NO generation for photoreceptor-mediated plant root growth regulation. Nitric Oxide 2019; 92:34-40. [PMID: 31377229 DOI: 10.1016/j.niox.2019.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023]
Abstract
Nitric oxide (NO) is an essential redox-signaling molecule free radical, contributes a significant role in a diverse range of physiological processes. Photo-triggered NO donors have significant potential compared to other NO donors because it releases NO in the presence of light. Hence, an efficient visible light-triggered NO donor is designed and synthesized by coupling 2,6-dimethyl nitrobenzene moiety at the peri-position of 1, 8-naphthalimide. The NO-releasing ability is validated using various spectroscopic techniques, the photoproduct is characterized, and finally, the NO generation quantum yield is also determined. Furthermore, the photo-generated NO has been employed to Arabidopsis thaliana as a model plant to examine the effect of photoreceptor-mediated NO uptake on plant root growth regulation molecule.
Collapse
|
4
|
Liu T, Zhang M, Mukosera GT, Borchardt D, Li Q, Tipple TE, Ishtiaq Ahmed AS, Power GG, Blood AB. L-NAME releases nitric oxide and potentiates subsequent nitroglycerin-mediated vasodilation. Redox Biol 2019; 26:101238. [PMID: 31200239 PMCID: PMC6565607 DOI: 10.1016/j.redox.2019.101238] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 12/22/2022] Open
Abstract
L-NG-Nitro arginine methyl ester (L-NAME) has been widely applied for several decades in both basic and clinical research as an antagonist of nitric oxide synthase (NOS). Herein, we show that L-NAME slowly releases NO from its guanidino nitro group. Daily pretreatment of rats with L-NAME potentiated mesenteric vasodilation induced by nitrodilators such as nitroglycerin, but not by NO. Release of NO also occurred with the NOS-inactive enantiomer D-NAME, but not with L-arginine or another NOS inhibitor L-NMMA, consistent with the presence or absence of a nitro group in their structure and their nitrodilator-potentiating effects. Metabolic conversion of the nitro group to NO-related breakdown products was confirmed using isotopically-labeled L-NAME. Consistent with Fenton chemistry, transition metals and reactive oxygen species accelerated the release of NO from L-NAME. Both NO production from L-NAME and its nitrodilator-potentiating effects were augmented under inflammation. NO release by L-NAME can confound its intended NOS-inhibiting effects, possibly by contributing to a putative intracellular NO store in the vasculature.
NOS-inhibitor L-NAME is also a precursor of NO. ROS releases NO from the nitro group of L-NAME via Fenton Chemistry. L-NAME potentates nitrodilator-mediated vasodilation. Nitroglycerin may cause vasodilation via activation of an intracellular NO store.
Collapse
Affiliation(s)
- Taiming Liu
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Meijuan Zhang
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - George T Mukosera
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Dan Borchardt
- Department of Chemistry, University of California, Riverside, CA, 92521, USA
| | - Qian Li
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Gordon G Power
- Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Arlin B Blood
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
5
|
Biswas S, Upadhyay N, Kar D, Datta S, Koner AL. Visible light-triggered NO generation from Naphthalimide-based probe for photoreceptor-mediated plant root growth regulation.. [DOI: 10.1101/550004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
Abstract
ABSTRACTAn efficient visible light-triggered nitric oxide (NO) releasing fluorescent molecule is designed and synthesized by coupling 2,6-dimethyl nitrobenzene moiety at the peri-position of 1, 8-naphthalimide through an alkene bond. The NO-releasing ability is investigated in details using various spectroscopic techniques, and the photoproduct was also characterized. Further, the photo-generated NO has been employed to examine the effect of photoreceptor-mediated NO uptake on plant root growth regulation.
Collapse
|
6
|
Ng TL, Rohac R, Mitchell AJ, Boal AK, Balskus EP. An N-nitrosating metalloenzyme constructs the pharmacophore of streptozotocin. Nature 2019; 566:94-99. [PMID: 30728519 PMCID: PMC6369591 DOI: 10.1038/s41586-019-0894-z] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022]
Abstract
Small molecules containing the N-nitroso group, such as the bacterial natural product streptozotocin, are prominent carcinogens1,2 and important cancer chemotherapeutics3,4. Despite the considerable importance of this functional group to human health, enzymes dedicated to the assembly of the N-nitroso unit have not been identified. Here we show that SznF, a metalloenzyme from the biosynthesis of streptozotocin, catalyses an oxidative rearrangement of the guanidine group of Nω-methyl-L-arginine to generate an N-nitrosourea product. Structural characterization and mutagenesis of SznF reveal two separate active sites that promote distinct steps in this transformation using different iron-containing metallocofactors. This biosynthetic reaction, which has little precedent in enzymology or organic synthesis, expands the catalytic capabilities of non-haem-iron-dependent enzymes to include N-N bond formation. We find that biosynthetic gene clusters that encode SznF homologues are widely distributed among bacteria-including environmental organisms, plant symbionts and human pathogens-which suggests an unexpectedly diverse and uncharacterized microbial reservoir of bioactive N-nitroso metabolites.
Collapse
Affiliation(s)
- Tai L Ng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Roman Rohac
- Department of Chemistry, The Pennsylvania State University, University Park, PA, USA
| | - Andrew J Mitchell
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Amie K Boal
- Department of Chemistry, The Pennsylvania State University, University Park, PA, USA.
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA.
| | - Emily P Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
7
|
González R, Molina-Ruiz FJ, Bárcena JA, Padilla CA, Muntané J. Regulation of Cell Survival, Apoptosis, and Epithelial-to-Mesenchymal Transition by Nitric Oxide-Dependent Post-Translational Modifications. Antioxid Redox Signal 2018; 29:1312-1332. [PMID: 28795583 DOI: 10.1089/ars.2017.7072] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Nitric oxide (NO) is a physiopathological messenger generating different reactive nitrogen species (RNS) according to hypoxic, acidic and redox conditions. Recent Advances: RNS and reactive oxygen species (ROS) promote relevant post-translational modifications, such as nitrosation, nitration, and oxidation, in critical components of cell proliferation and death, epithelial-to-mesenchymal transition, and metastasis. CRITICAL ISSUES The pro- or antitumoral properties of NO are dependent on local concentration, redox state, cellular status, duration of exposure, and compartmentalization of NO generation. The increased expression of NO synthase has been associated with cancer progression. However, the experimental strategies leading to high intratumoral NO generation have been shown to exert antitumoral properties. The effect of NO and ROS on cell signaling is critically altered by factors modulating tumor progression such as oxygen content, metabolism, and inflammatory response. The review describes the alteration of key components involved in cell survival and death, metabolism, and metastasis induced by RNS- and ROS-related post-translational modifications. FUTURE DIRECTIONS The identification of the molecular targets affected by nitrosation, nitration, and oxidation, as well as their interactions with other post-translational modifications, will improve the understanding on the complex signaling and cell fate decision in cancer. The therapeutic NO-based strategies have to address the complex crosstalk among NO and ROS with regard to critical components affecting tumor cell survival, metabolism, and metastasis in the progression of cancer, as well as close interaction with ionizing radiation and chemotherapy.
Collapse
Affiliation(s)
- Raúl González
- 1 Institute of Biomedicine of Seville (IBiS), IBiS/"Virgen del Rocío" University Hospital/CSIC/University of Seville , Seville, Spain
| | - Francisco J Molina-Ruiz
- 1 Institute of Biomedicine of Seville (IBiS), IBiS/"Virgen del Rocío" University Hospital/CSIC/University of Seville , Seville, Spain
| | - J Antonio Bárcena
- 2 Department of Biochemistry and Molecular Biology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba , Córdoba, Spain
| | - C Alicia Padilla
- 2 Department of Biochemistry and Molecular Biology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba , Córdoba, Spain
| | - Jordi Muntané
- 3 Department of General Surgery, "Virgen del Rocío" University Hospital/IBiS/CSIC/University of Seville , Seville, Spain .,4 Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) , Madrid, Spain
| |
Collapse
|
8
|
González-Montero J, Brito R, Gajardo AIJ, Rodrigo R. Myocardial reperfusion injury and oxidative stress: Therapeutic opportunities. World J Cardiol 2018; 10:74-86. [PMID: 30344955 PMCID: PMC6189069 DOI: 10.4330/wjc.v10.i9.74] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/27/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of death worldwide. Its associated mortality, morbidity and complications have significantly decreased with the development of interventional cardiology and percutaneous coronary angioplasty (PCA) treatment, which quickly and effectively restore the blood flow to the area previously subjected to ischemia. Paradoxically, the restoration of blood flow to the ischemic zone leads to a massive production of reactive oxygen species (ROS) which generate rapid and severe damage to biomolecules, generating a phenomenon called myocardial reperfusion injury (MRI). In the clinical setting, MRI is associated with multiple complications such as lethal reperfusion, no-reflow, myocardial stunning, and reperfusion arrhythmias. Despite significant advances in the understanding of the mechanisms accounting for the myocardial ischemia reperfusion injury, it remains an unsolved problem. Although promising results have been obtained in experimental studies (mainly in animal models), these benefits have not been translated into clinical settings. Thus, clinical trials have failed to find benefits from any therapy to prevent MRI. There is major evidence with respect to the contribution of oxidative stress to MRI in cardiovascular diseases. The lack of consistency between basic studies and clinical trials is not solely based on the diversity inherent in epidemiology but is also a result of the methodological weaknesses of some studies. It is quite possible that pharmacological issues, such as doses, active ingredients, bioavailability, routes of administration, co-therapies, startup time of the drug intervention, and its continuity may also have some responsibility for the lack of consistency between different studies. Furthermore, the administration of high ascorbate doses prior to reperfusion appears to be a safe and rational therapy against the development of oxidative damage associated with myocardial reperfusion. In addition, the association with N-acetylcysteine (a glutathione donor) and deferoxamine (an iron chelator) could improve the antioxidant cardioprotection by ascorbate, making it even more effective in preventing myocardial reperfusion damage associated with PCA following AMI.
Collapse
Affiliation(s)
- Jaime González-Montero
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| | - Roberto Brito
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Internal Medicine Department, University of Chile, Clinical Hospital, Santiago 70058, Chile
| | - Abraham IJ Gajardo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
- Internal Medicine Department, University of Chile, Clinical Hospital, Santiago 70058, Chile
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 70058, Chile
| |
Collapse
|
9
|
Cao GJ, Fisher CM, Jiang X, Chong Y, Zhang H, Guo H, Zhang Q, Zheng J, Knolhoff AM, Croley TR, Yin JJ. Platinum nanoparticles: an avenue for enhancing the release of nitric oxide from S-nitroso-N-acetylpenicillamine and S-nitrosoglutathione. NANOSCALE 2018; 10:11176-11185. [PMID: 29873378 DOI: 10.1039/c8nr03874k] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nitric oxide (NO) is an endogenous bioregulator with established roles in diverse fields. The difficulty in the modulation of NO release is still a significant obstacle to achieving successful clinical applications. We report herein our initial work using electron spin resonance (ESR) spectroscopy to detect NO generated from S-nitroso-N-acetylpenicillamine (SNAP) and S-nitrosoglutathione (GSNO) donors catalyzed by platinum nanoparticles (Pt NPs, 3 nm) under physiological conditions. With ESR spectroscopy coupled with spin trapping and spin labeling techniques, we identified that Pt NPs can significantly promote the generation of NO from SNAP and GSNO under physiological conditions. A classic NO colorimetric detection kit was also employed to verify that Pt NPs truly triggered the release of NO from its donors. Pt NPs can act as promising delivery vehicles for on-demand NO delivery based on time and dosage. These results, along with the detection of the resulting disulfide product, were confirmed with mass spectrometry. In addition, cellular experiments provided a convincing demonstration that the triggered release of NO from its donors by Pt NPs is efficient in killing human cancer cells in vitro. The catalytic mechanism was elucidated by X-ray photo-electron spectroscopy (XPS) and ultra-high performance liquid chromatography/high-resolution mass spectrometry (UHPLC-HRMS), which suggested that Pt-S bond formation occurs in the solution of Pt NPs and NO donors. Identification of Pt NPs capable of generating NO from S-nitrosothiols (RSNOs) is an important step in harnessing NO for investigations into its clinical applications and therapies.
Collapse
Affiliation(s)
- Gao-Juan Cao
- Department of Applied Chemistry, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Hardy M, Zielonka J, Karoui H, Sikora A, Michalski R, Podsiadły R, Lopez M, Vasquez-Vivar J, Kalyanaraman B, Ouari O. Detection and Characterization of Reactive Oxygen and Nitrogen Species in Biological Systems by Monitoring Species-Specific Products. Antioxid Redox Signal 2018; 28:1416-1432. [PMID: 29037049 PMCID: PMC5910052 DOI: 10.1089/ars.2017.7398] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 10/15/2017] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Since the discovery of the superoxide dismutase enzyme, the generation and fate of short-lived oxidizing, nitrosating, nitrating, and halogenating species in biological systems has been of great interest. Despite the significance of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in numerous diseases and intracellular signaling, the rigorous detection of ROS and RNS has remained a challenge. Recent Advances: Chemical characterization of the reactions of selected ROS and RNS with electron paramagnetic resonance (EPR) spin traps and fluorescent probes led to the establishment of species-specific products, which can be used for specific detection of several forms of ROS and RNS in cell-free systems and in cultured cells in vitro and in animals in vivo. Profiling oxidation products from the ROS and RNS probes provides a rigorous method for detection of those species in biological systems. CRITICAL ISSUES Formation and detection of species-specific products from the probes enables accurate characterization of the oxidative environment in cells. Measurement of the total signal (fluorescence, chemiluminescence, etc.) intensity does not allow for identification of the ROS/RNS formed. It is critical to identify the products formed by using chromatographic or other rigorous techniques. Product analyses should be accompanied by monitoring of the intracellular probe level, another factor controlling the yield of the product(s) formed. FUTURE DIRECTIONS More work is required to characterize the chemical reactivity of the ROS/RNS probes, and to develop new probes/detection approaches enabling real-time, selective monitoring of the specific products formed from the probes. Antioxid. Redox Signal. 28, 1416-1432.
Collapse
Affiliation(s)
- Micael Hardy
- Aix Marseille Univ, CNRS, ICR, Marseille, France
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hakim Karoui
- Aix Marseille Univ, CNRS, ICR, Marseille, France
| | - Adam Sikora
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Lodz, Poland
| | - Radosław Michalski
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Lodz, Poland
| | - Radosław Podsiadły
- Faculty of Chemistry, Institute of Polymer and Dye Technology, Lodz University of Technology, Lodz, Poland
| | - Marcos Lopez
- Translational Biomedical Research Group, Biotechnology Laboratories, Cardiovascular Foundation of Colombia, Santander, Colombia
- Graduate Program of Biomedical Sciences, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
11
|
|
12
|
Detection of Nitric Oxide by Electron Paramagnetic Resonance Spectroscopy: Spin-Trapping with Iron-Dithiocarbamates. Methods Mol Biol 2016; 1424:81-102. [PMID: 27094413 DOI: 10.1007/978-1-4939-3600-7_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Electron paramagnetic resonance (EPR) spectroscopy is the ideal methodology to identify radicals (detection and characterization of molecular structure) and to study their kinetics, in both simple and complex biological systems. The very low concentration and short life-time of NO and of many other radicals do not favor its direct detection and spin-traps are needed to produce a new and persistent radical that can be subsequently detected by EPR spectroscopy.In this chapter, we present the basic concepts of EPR spectroscopy and of some spin-trapping methodologies to study NO. The "strengths and weaknesses" of iron-dithiocarbamates utilization, the NO traps of choice for the authors, are thoroughly discussed and a detailed description of the method to quantify the NO formation by molybdoenzymes is provided.
Collapse
|
13
|
Sydow K, Hornig B, Arakawa N, Bode-Böger SM, Tsikas D, Münzel T, Böger RH. Endothelial dysfunction in patients with peripheral arterial disease and chronic hyperhomocysteinemia: potential role of ADMA. Vasc Med 2016; 9:93-101. [PMID: 15521698 DOI: 10.1191/1358863x04vm538oa] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hyperhomocysteinemia is associated with an enhanced risk for cardiovascular disease. Patients with peripheral arterial disease (PAD) show an increased prevalence of hyper-homocysteinemia. A decreased biological activity of nitric oxide (NO) may contribute to homocysteine-associated endothelial dysfunction. This study was designed to investigate whether elevated levels of the endogenous NO synthase inhibitor asymmetric dimethylarginine (ADMA) are involved in endothelial dysfunction in patients with chronic hyperhomocysteinemia and PAD. A total of 76 patients (58 males and 18 females; mean age 65.2 2.0 years) with PAD were included in the analysis and characterized according to demographic variables and cardiovascular risk factors. Flow-dependent vasodilation (FDD) was determined by high-resolution ultrasound in the radial artery. Total plasma homocysteine (plasma tHcy) and ADMA levels were measured by HPLC. Urinary nitrate was quantified using gas chromatography-mass spectrometry. Patients with plasma tHcy in the highest tertile (n 1/4 27; i.e. >10.6 mmol=l) had a mean plasma level of 14.4 1.2 mmol=l compared with 9.9 0.1 mmol=l in those patients in the middle tertile (n 1/4 22; p < 0.05) and 9.4 0.1 mmol=l in those in the lowest tertile (n 1/4 27; i.e. <9.6 mmol=l; p < 0.05). The hyperhomocysteinemic individuals (highest tertile) had a significantly decreased FDD compared with healthy age-matched controls (n 1/4 15) (7.6 1.0 vs 13.0 0.4%; p < 0.05), higher plasma ADMA concentrations (4.0 0.3 vs 2.6 0.3 mmol=l; p < 0.05), and a lower urinary nitrate excretion rate (89.5 13.4 vs 131.3 17.9 mmol=mmol creatinine; p < 0.05) compared with patients with plasma tHcy in the lowest tertile. Multivariate regression analysis including plasma tHcy, ADMA, total cholesterol, diabetes mellitus, smoking, and systolic blood pressure revealed ADMA as the only significant factor determining FDD (p < 0.05). In conclusion, we demonstrated a stronger relationship between impaired endothelial function and elevated ADMA levels in comparison with plasma tHcy concentrations in patients with PAD and chronic hyperhomocysteinemia. This may raise the question of whether different therapeutical options that interact indirectly with plasma tHcy, i.e. treatment with ACE inhibitors and AT1-receptor blockers to reduce ADMA plasma concentrations or L-arginine, could be a beneficial tool for treating patients with hyperhomocysteinemia.
Collapse
Affiliation(s)
- Karsten Sydow
- Division of Cardiology, University Hospital Hamburg-Eppendorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
Hirsh DJ, Schieler BM, Fomchenko KM, Jordan ET, Bidle KD. A liposome-encapsulated spin trap for the detection of nitric oxide. Free Radic Biol Med 2016; 96:199-210. [PMID: 27112665 DOI: 10.1016/j.freeradbiomed.2016.04.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/09/2016] [Accepted: 04/21/2016] [Indexed: 01/08/2023]
Abstract
Electron paramagnetic resonance (EPR) is one of the few methods that allows for the unambiguous detection of nitric oxide (NO). However, the dithiocarbamate-iron spin traps employed with this method inhibit the activity of nitric oxide synthase and catalyze NO production from nitrite. These disadvantages limit EPR's application to biological NO detection. We present a liposome-encapsulated spin-trap (LEST) method for the capture and in situ detection of NO by EPR. The method shows a linear response for [NO]≥4µM and can detect [NO]≥40nM in a 500µL sample (≥20 pmol). The kinetics of NO production can be followed in real time over minutes to hours. LEST does not inhibit the activity of inducible nitric oxide synthase or nitrate reductase and shows minimal abiotic NO production in the presence of nitrite and NADH. Nitrate reductase-like activity is detected in cell lysates of the coccolithophore Emiliania huxleyi and is elevated in virus-infected culture. This method shows particular promise for NO detection in cell lysates and crude preparations of NO-producing tissues.
Collapse
Affiliation(s)
- Donald J Hirsh
- Department of Chemistry, The College of New Jersey, Ewing, NJ 08628, United States
| | - Brittany M Schieler
- Department of Marine and Coastal Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | | | - Ethan T Jordan
- Department of Biology, Marine Biology & Environmental Science, William Rogers University, Bristol, RI 02809, United States
| | - Kay D Bidle
- Department of Marine and Coastal Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| |
Collapse
|
15
|
Casas AI, Dao VTV, Daiber A, Maghzal GJ, Di Lisa F, Kaludercic N, Leach S, Cuadrado A, Jaquet V, Seredenina T, Krause KH, López MG, Stocker R, Ghezzi P, Schmidt HHHW. Reactive Oxygen-Related Diseases: Therapeutic Targets and Emerging Clinical Indications. Antioxid Redox Signal 2015; 23:1171-85. [PMID: 26583264 PMCID: PMC4657512 DOI: 10.1089/ars.2015.6433] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Enhanced levels of reactive oxygen species (ROS) have been associated with different disease states. Most attempts to validate and exploit these associations by chronic antioxidant therapies have provided disappointing results. Hence, the clinical relevance of ROS is still largely unclear. RECENT ADVANCES We are now beginning to understand the reasons for these failures, which reside in the many important physiological roles of ROS in cell signaling. To exploit ROS therapeutically, it would be essential to define and treat the disease-relevant ROS at the right moment and leave physiological ROS formation intact. This breakthrough seems now within reach. CRITICAL ISSUES Rather than antioxidants, a new generation of protein targets for classical pharmacological agents includes ROS-forming or toxifying enzymes or proteins that are oxidatively damaged and can be functionally repaired. FUTURE DIRECTIONS Linking these target proteins in future to specific disease states and providing in each case proof of principle will be essential for translating the oxidative stress concept into the clinic.
Collapse
Affiliation(s)
- Ana I Casas
- 1 Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - V Thao-Vi Dao
- 1 Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - Andreas Daiber
- 2 2nd Medical Department, Molecular Cardiology, University Medical Center , Mainz, Germany
| | - Ghassan J Maghzal
- 3 Victor Chang Cardiac Research Institute, and School of Medical Sciences, University of New South Wales , Sydney, New South Wales, Australia
| | - Fabio Di Lisa
- 4 Department of Biomedical Sciences, University of Padova , Italy .,5 Neuroscience Institute , CNR, Padova, Italy
| | | | - Sonia Leach
- 6 Brighton and Sussex Medical School , Falmer, United Kingdom
| | - Antonio Cuadrado
- 7 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid , Madrid, Spain
| | - Vincent Jaquet
- 8 Department of Pathology and Immunology, Medical School, University of Geneva , Geneva, Switzerland
| | - Tamara Seredenina
- 8 Department of Pathology and Immunology, Medical School, University of Geneva , Geneva, Switzerland
| | - Karl H Krause
- 8 Department of Pathology and Immunology, Medical School, University of Geneva , Geneva, Switzerland
| | - Manuela G López
- 9 Teofilo Hernando Institute, Department of Pharmacology, Faculty of Medicine. Autonomous University of Madrid , Madrid, Spain
| | - Roland Stocker
- 3 Victor Chang Cardiac Research Institute, and School of Medical Sciences, University of New South Wales , Sydney, New South Wales, Australia
| | - Pietro Ghezzi
- 6 Brighton and Sussex Medical School , Falmer, United Kingdom
| | - Harald H H W Schmidt
- 1 Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| |
Collapse
|
16
|
Jiang H, Shen Z, Chu Y, Li Y, Li J, Wang X, Yang W, Zhang X, Ju J, Xu J, Yang C. Serum metabolomics research of the anti-hypertensive effects of Tengfu Jiangya tablet on spontaneously hypertensive rats. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 1002:210-7. [DOI: 10.1016/j.jchromb.2015.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 06/15/2015] [Accepted: 08/07/2015] [Indexed: 01/05/2023]
|
17
|
Ischaemic conditioning strategies reduce ischaemia/reperfusion-induced organ injury. Br J Anaesth 2015; 114:204-16. [DOI: 10.1093/bja/aeu302] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
18
|
Maia LB, Pereira V, Mira L, Moura JJG. Nitrite reductase activity of rat and human xanthine oxidase, xanthine dehydrogenase, and aldehyde oxidase: evaluation of their contribution to NO formation in vivo. Biochemistry 2015; 54:685-710. [PMID: 25537183 DOI: 10.1021/bi500987w] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitrite is presently considered a NO "storage form" that can be made available, through its one-electron reduction, to maintain NO formation under hypoxia/anoxia. The molybdoenzymes xanthine oxidase/dehydrogenase (XO/XD) and aldehyde oxidase (AO) are two of the most promising mammalian nitrite reductases, and in this work, we characterized NO formation by rat and human XO/XD and AO. This is the first characterization of human enzymes, and our results support the employment of rat liver enzymes as suitable models of the human counterparts. A comprehensive kinetic characterization of the effect of pH on XO and AO-catalyzed nitrite reduction showed that the enzyme's specificity constant for nitrite increase 8-fold, while the Km(NO2(-)) decrease 6-fold, when the pH decreases from 7.4 to 6.3. These results demonstrate that the ability of XO/AO to trigger NO formation would be greatly enhanced under the acidic conditions characteristic of ischemia. The dioxygen inhibition was quantified, and the Ki(O2) values found (24.3-48.8 μM) suggest that in vivo NO formation would be fine-tuned by dioxygen availability. The potential in vivo relative physiological relevance of XO/XD/AO-dependent pathways of NO formation was evaluated using HepG2 and HMEC cell lines subjected to hypoxia. NO formation by the cells was found to be pH-, nitrite-, and dioxygen-dependent, and the relative contribution of XO/XD plus AO was found to be as high as 50%. Collectively, our results supported the possibility that XO/XD and AO can contribute to NO generation under hypoxia inside a living human cell. Furthermore, the molecular mechanism of XO/AO-catalyzed nitrite reduction was revised.
Collapse
Affiliation(s)
- Luisa B Maia
- UCIBIO, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa , 2829-516 Caparica, Portugal
| | | | | | | |
Collapse
|
19
|
Das A, Gopalakrishnan B, Druhan LJ, Wang TY, De Pascali F, Rockenbauer A, Racoma I, Varadharaj S, Zweier JL, Cardounel AJ, Villamena FA. Reversal of SIN-1-induced eNOS dysfunction by the spin trap, DMPO, in bovine aortic endothelial cells via eNOS phosphorylation. Br J Pharmacol 2014; 171:2321-34. [PMID: 24405159 DOI: 10.1111/bph.12572] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/03/2013] [Accepted: 12/18/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Nitric oxide (NO) derived from eNOS is mostly responsible for the maintenance of vascular homeostasis and its decreased bioavailability is characteristic of reactive oxygen species (ROS)-induced endothelial dysfunction (ED). Because 5,5-dimethyl-1-pyrroline-N-oxide (DMPO), a commonly used spin trap, can control intracellular nitroso-redox balance by scavenging ROS and donating NO, it was employed as a cardioprotective agent against ED but the mechanism of its protection is still not clear. This study elucidated the mechanism of protection by DMPO against SIN-1-induced oxidative injury to bovine aortic endothelial cells (BAEC). EXPERIMENTAL APPROACH BAEC were treated with SIN-1, as a source of peroxynitrite anion (ONOO⁻), and then incubated with DMPO. Cytotoxicity following SIN-1 alone and cytoprotection by adding DMPO was assessed by MTT assay. Levels of ROS and NO generation from HEK293 cells transfected with wild-type and mutant eNOS cDNAs, tetrahydrobiopterin bioavailability, eNOS activity, eNOS and Akt kinase phosphorylation were measured. KEY RESULTS Post-treatment of cells with DMPO attenuated SIN-1-mediated cytotoxicity and ROS generation, restoration of NO levels via increased in eNOS activity and phospho-eNOS levels. Treatment with DMPO alone significantly increased NO levels and induced phosphorylation of eNOS Ser¹¹⁷⁹ via Akt kinase. Transfection studies with wild-type and mutant human eNOS confirmed the dual role of eNOS as a producer of superoxide anion (O₂⁻) with SIN-1 treatment, and a producer of NO in the presence of DMPO. CONCLUSION AND IMPLICATIONS Post-treatment with DMPO of oxidatively challenged cells reversed eNOS dysfunction and could have pharmacological implications in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Amlan Das
- Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Trescher K, Dzilic E, Kreibich M, Gasser H, Aumayr K, Kerjaschki D, Pelzmann B, Hallström S, Podesser BK. The nitric oxide donor, S-nitroso human serum albumin, as an adjunct to HTK-N cardioplegia improves protection during cardioplegic arrest after myocardial infarction in rats. Interact Cardiovasc Thorac Surg 2014; 20:387-94. [PMID: 25468794 DOI: 10.1093/icvts/ivu383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Currently available cardioplegic solutions provide excellent protection in patients with normal surgical risk; in high-risk patients, however, such as in emergency coronary artery bypass surgery, there is still room for improvement. As most of the cardioplegic solutions primarily protect myocytes, the addition of substances for protection of the endothelium might improve their protective potential. The nitric oxide donor, S-nitroso human serum albumin (S-NO-HSA), which has been shown to prevent endothelial nitric oxide synthase uncoupling, was added to the newly developed histidine-tryptophan-ketoglutarat (HTK-N) cardioplegia in an isolated heart perfusion system after subjecting rats to acute myocardial infarction (MI) and reperfusion. METHODS In male Sprague-Dawley rats, acute MI was induced by ligation for 1 h of the anterior descending coronary artery. After 2 h of in vivo reperfusion hearts were evaluated on an isolated erythrocyte-perfused working heart model. Cold ischaemia (4°C) for 60 min was followed by 45 min of reperfusion. Cardiac arrest was induced either with HTK (n = 10), HTK-N (n = 10) or HTK-N + S-NO-HSA (n = 10). In one group (HTK-N + S-NO-HSA plus in vivo S-NO-HSA; n = 9) an additional in vivo infusion of S-NO-HSA was performed. RESULTS Post-ischaemic recovery of cardiac output (HTK: 77 ± 4%, HTK-N: 86 ± 7%, HTK-N + S-NO-HSA: 101 ± 5%, in vivo S-NO-HSA: 93 ± 8%), external heart work (HTK: 79 ± 5%, HTK-N: 83 ± 3%, HTK-N + S-NO-HSA: 101 ± 8%, in vivo S-NO-HSA: 109 ± 13%), coronary flow (HTK: 77 ± 4%, HTK-N: 94 ± 6%, HTK-N + S-NO-HSA: 118 ± 15%, in vivo S-NO-HSA: 113 ± 3.17%) [HTK-N + S-NO-HSA vs HTK P < 0.001; HTK-N + S-NO-HSA vs HTK-N P < 0.05] and left atrial diastolic pressure (HTK: 122 ± 31%, HTK-N: 159 ± 43%, HTK-N + S-NO-HSA: 88 ± 30, in vivo S-NO-HSA: 62 ± 10%) [HTK-N + S-NO-HSA vs HTK P < 0.05; in vivo S-NO-HSA vs HTK-N P < 0.05] were significantly improved in both S-NO-HSA-treated groups compared with HTK and HTK-N, respectively. This was accompanied by better preservation of high-energy phosphates (adenosine triphosphate; energy charge) and ultrastructural integrity on transmission electron microscopy. However, no additional benefit of in vivo S-NO-HSA infusion was observed. CONCLUSIONS Addition of the NO donor, S-NO-HSA refines the concept of HTK-N cardioplegia in improving post-ischaemic myocardial perfusion. HTK-N with S-NO-HSA is a possible therapeutic option for patients who have to be operated on for acute MI.
Collapse
Affiliation(s)
- Karola Trescher
- Department of Cardiac Surgery, LK St. Pölten, Pölten, Austria Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna Medical University, Vienna, Austria
| | - Elda Dzilic
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna Medical University, Vienna, Austria
| | - Maximilian Kreibich
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna Medical University, Vienna, Austria
| | - Harald Gasser
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna Medical University, Vienna, Austria
| | - Klaus Aumayr
- Department of Pathology, Vienna Medical University, Vienna, Austria
| | | | - Brigitte Pelzmann
- Institute of Biophysics, Center of Physiological Medicine, Medical University Graz, Graz, Austria
| | - Seth Hallström
- Institute of Physiological Chemistry, Center of Physiological Medicine, Medical University Graz, Graz, Austria
| | - Bruno K Podesser
- Department of Cardiac Surgery, LK St. Pölten, Pölten, Austria Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna Medical University, Vienna, Austria
| |
Collapse
|
21
|
Rodrigo R, Libuy M, Feliú F, Hasson D. Oxidative stress-related biomarkers in essential hypertension and ischemia-reperfusion myocardial damage. DISEASE MARKERS 2013; 35:773-90. [PMID: 24347798 PMCID: PMC3856219 DOI: 10.1155/2013/974358] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022]
Abstract
Cardiovascular diseases are a leading cause of mortality and morbidity worldwide, with hypertension being a major risk factor. Numerous studies support the contribution of reactive oxygen and nitrogen species in the pathogenesis of hypertension, as well as other pathologies associated with ischemia/reperfusion. However, the validation of oxidative stress-related biomarkers in these settings is still lacking and novel association of these biomarkers and other biomarkers such as endothelial progenitor cells, endothelial microparticles, and ischemia modified albumin, is just emerging. Oxidative stress has been suggested as a pathogenic factor and therapeutic target in early stages of essential hypertension. Systolic and diastolic blood pressure correlated positively with plasma F2-isoprostane levels and negatively with total antioxidant capacity of plasma in hypertensive and normotensive patients. Cardiac surgery with extracorporeal circulation causes an ischemia/reperfusion event associated with increased lipid peroxidation and protein carbonylation, two biomarkers associated with oxidative damage of cardiac tissue. An enhancement of the antioxidant defense system should contribute to ameliorating functional and structural abnormalities derived from this metabolic impairment. However, data have to be validated with the analysis of the appropriate oxidative stress and/or nitrosative stress biomarkers.
Collapse
Affiliation(s)
- Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Matías Libuy
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Felipe Feliú
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| | - Daniel Hasson
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Independencia 1027, Casilla 70058, Santiago 7, Chile
| |
Collapse
|
22
|
Karki R, Park CH, Kim DW. Extract of buckwheat sprouts scavenges oxidation and inhibits pro-inflammatory mediators in lipopolysaccharide-stimulated macrophages (RAW264.7). JOURNAL OF INTEGRATIVE MEDICINE-JIM 2013; 11:246-52. [PMID: 23867243 DOI: 10.3736/jintegrmed2013036] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Buckwheat has been considered as a potential source of nutraceutical components on the world market of probiotic foodstuffs. The purpose of this study was to evaluate the effects of tartary buckwheat (Fagopyrum tataricum) sprouts on oxidation and pro-inflammatory mediators. METHODS The anti-oxidant effects of buckwheat extract (BWE) and rutin were evaluated by using 1,1-diphenyl-2-picrylhydrazyl (DPPH)- and nitric oxide (NO)-scavenging activities, serum peroxidation and chelating assays. Lipopolysaccharide (LPS)-stimulated RAW264.7 cells were used to evaluate anti-inflammatory activities of buckwheat and rutin. NO production in LPS-stimulated RAW264.7 cells was determined by using Griess reagent. The expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nuclear factor-kappa B (NF-κB) p65 subunit in cytosolic and nuclear portions were determined by Western blot analysis. Also, the production of inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) was determined by enzyme-linked immunosorbent assay. RESULTS Inhibitory concentration 50 values for DPPH- and NO-scavenging activities of BWE were 24.97 and 72.54 μg/mL respectively. BWE inhibited serum oxidation and possessed chelating activity. Furthermore, BWE inhibited IL-6 and TNF-α production in LPS-stimulated RAW264.7 cells. Also, BWE inhibited iNOS and COX-2 expression and NF-κB p65 translocation. CONCLUSION Buckwheat sprouts possessed strong antioxidant activity and inhibited production of pro-inflammatory mediators in the applied model systems. Thus, buckwheat can be suggested to be beneficial in inflammatory diseases by inhibiting the free radicals and inflammatory mediators.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Oriental Medicine Resources, Mokpo National University, Muan-gun, Jeollanam-do 534-729, South Korea; E-mail: ,
| | | | | |
Collapse
|
23
|
Knott ME, Dorfman D, Chianelli MS, Sáenz DA. Effect of Angeli’s salt on the glutamate/glutamine cycle activity and on glutamate excitotoxicity in the hamster retina. Neurochem Int 2012; 61:7-15. [DOI: 10.1016/j.neuint.2012.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 03/30/2012] [Accepted: 04/02/2012] [Indexed: 10/28/2022]
|
24
|
Aoki C, Nakano A, Tanaka S, Yanagi K, Ohta S, Jojima T, Kasai K, Takekawa H, Hirata K, Hattori Y. Fluvastatin upregulates endothelial nitric oxide synthase activity via enhancement of its phosphorylation and expression and via an increase in tetrahydrobiopterin in vascular endothelial cells. Int J Cardiol 2012; 156:55-61. [PMID: 21093076 DOI: 10.1016/j.ijcard.2010.10.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 08/17/2010] [Accepted: 10/23/2010] [Indexed: 11/22/2022]
Abstract
BACKGROUND An HMG-CoA reductase inhibitor, fluvastatin, appears to act directly on the blood vessel wall to stabilize plaques in situ, agents that share this property have been termed vascular statins. METHODS We investigated the effects of fluvastatin on endothelial nitric oxide synthase (eNOS) phosphorylation and expression, as well as terahydrobiopterin (BH4) metabolism, in human umbilical vein endothelial cells (HUVEC). RESULTS Fluvastatin was observed to enhance eNOS phosphorylation at Ser-1177 and Ser-633 through the PI3-kinase/Akt and PKA pathways, respectively. Inhibition of eNOS phosphorylation using inhibitors of these pathways attenuated acute NO release in response to fluvastatin. The mRNA of GTP cyclohydrolase I (GTPCH), the rate-limiting enzyme of the first step of de novo BH4 synthesis, as well as eNOS, was upregulated in HUVEC treated with fluvastatin. In parallel with this observation, fluvastatin increased intracellular BH4. Pre-treatment of HUVEC with the selective GTPCH inhibitor, 2,4-diamino-6-hydroxypyrimidine, reduced intracellular BH4 and decreased citrulline formation following stimulation with ionomycin. Furthermore, the potentiating effect of fluvastatin was reduced by limiting the cellular availability of BH4. CONCLUSIONS Our data demonstrate that fluvastatin phosphorylates and activates eNOS, and increases eNOS expression in vascular endothelial cells. In addition to modulating eNOS, fluvastatin potentiates GTPCH gene expression and BH4 synthesis, thereby increasing NO production and preventing relative shortages of BH4.
Collapse
Affiliation(s)
- Chie Aoki
- Department of Endocrinology and Metabolism, Dokkyo University School of Medicine, Mibu, Tochigi 321-0293, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Álvares TS, Conte CA, Paschoalin VMF, Silva JT, Meirelles CDM, Bhambhani YN, Gomes PSC. Acute l-arginine supplementation increases muscle blood volume but not strength performance. Appl Physiol Nutr Metab 2012; 37:115-26. [DOI: 10.1139/h11-144] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
l-Arginine (L-arg) is an amino acid precursor to nitric oxide (NO). Dietary supplements containing L-arg have been marketed with the purpose of increasing vasodilation, thereby elevating blood flow to the exercising muscle and enhancing the metabolic response to exercise. Our goal was to identify the acute effect of L-arg supplementation on biceps strength performance, indicators of NO production (nitrite and nitrate – NOx), and muscle blood volume (Mbv) and oxygenation (Mox) during recovery from 3 sets of resistance exercise. Fifteen males participated in a randomized, double-blind, placebo-controlled study. After withdrawing resting blood samples, the subjects were supplemented with 6 g of L-arg (ARG) or placebo (PLA). Monitoring of Mbv and Mox with near-infrared spectroscopy began 30 min after supplementation and lasted for 60 min. The exercise protocol (3 sets of 10 maximal voluntary contractions of isokinetic concentric elbow extension at 60°·s–1, 2-min rest between sets) was initiated 80 min after supplementation. Blood samples were drawn at 30, 60, 90, and 120 min after supplementation. Repeated measures ANOVA showed that Mbv significantly (p ≤ 0.05) increased in ARG compared with the PLA during the recovery period of each set of resistance exercise. NOx, Mox, peak torque, total work, and set total work were not significantly different between groups. We found that acute L-arg supplementation increases Mbv during recovery from sets of resistance exercise with no increase in strength performance. It is still premature to recommend nutritional supplements containing L-arg as an ergogenic aid to increase muscle strength during resistance training in healthy subjects.
Collapse
Affiliation(s)
- Thiago Silveira Álvares
- Laboratory Crossbridges, Physical Education Postgraduate Program, Gama Filho University, Rio de Janeiro, Brazil
- Department of Biochemistry, Chemistry Institute, Federal University of Rio de Janeiro, Brazil
- Laboratório de Análises Avançadas em Bioquímica e Biologia Molecular, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos, 149 Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Carlos Adam Conte
- Department of Biochemistry, Chemistry Institute, Federal University of Rio de Janeiro, Brazil
| | | | - Joab Trajano Silva
- Department of Biochemistry, Chemistry Institute, Federal University of Rio de Janeiro, Brazil
| | - Cláudia de Mello Meirelles
- Laboratory Crossbridges, Physical Education Postgraduate Program, Gama Filho University, Rio de Janeiro, Brazil
- School of Physical Education of the Army, Rio de Janeiro, Brazil
| | - Yagesh N. Bhambhani
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, AB T6G 2G4, Canada
| | - Paulo Sergio Chagas Gomes
- Laboratory Crossbridges, Physical Education Postgraduate Program, Gama Filho University, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Jain M, Barthwal MK, Haq W, Katti SB, Dikshit M. Synthesis and Pharmacological Evaluation of Novel Arginine Analogs as Potential Inhibitors of Acetylcholine-Induced Relaxation in Rat Thoracic Aortic Rings. Chem Biol Drug Des 2012; 79:459-69. [DOI: 10.1111/j.1747-0285.2011.01286.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Raedschelders K, Ansley DM, Chen DDY. The cellular and molecular origin of reactive oxygen species generation during myocardial ischemia and reperfusion. Pharmacol Ther 2011; 133:230-55. [PMID: 22138603 DOI: 10.1016/j.pharmthera.2011.11.004] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/04/2011] [Indexed: 02/07/2023]
Abstract
Myocardial ischemia-reperfusion injury is an important cause of impaired heart function in the early postoperative period subsequent to cardiac surgery. Reactive oxygen species (ROS) generation increases during both ischemia and reperfusion and it plays a central role in the pathophysiology of intraoperative myocardial injury. Unfortunately, the cellular source of these ROS during ischemia and reperfusion is often poorly defined. Similarly, individual ROS members tend to be grouped together as free radicals with a uniform reactivity towards biomolecules and with deleterious effects collectively ascribed under the vague umbrella of oxidative stress. This review aims to clarify the identity, origin, and progression of ROS during myocardial ischemia and reperfusion. Additionally, this review aims to describe the biochemical reactions and cellular processes that are initiated by specific ROS that work in concert to ultimately yield the clinical manifestations of myocardial ischemia-reperfusion. Lastly, this review provides an overview of several key cardioprotective strategies that target myocardial ischemia-reperfusion injury from the perspective of ROS generation. This overview is illustrated with example clinical studies that have attempted to translate these strategies to reduce the severity of ischemia-reperfusion injury during coronary artery bypass grafting surgery.
Collapse
Affiliation(s)
- Koen Raedschelders
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine. The University of British Columbia, Vancouver, BC, Canada.
| | | | | |
Collapse
|
28
|
Nitrosyl hydride (HNO) replaces dioxygen in nitroxygenase activity of manganese quercetin dioxygenase. Proc Natl Acad Sci U S A 2011; 108:18926-31. [PMID: 22084064 DOI: 10.1073/pnas.1111488108] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Quercetin dioxygenase (QDO) catalyzes the oxidation of the flavonol quercetin with dioxygen, cleaving the central heterocyclic ring and releasing CO. The QDO from Bacillus subtilis is unusual in that it has been shown to be active with several divalent metal cofactors such as Fe, Mn, and Co. Previous comparison of the catalytic activities suggest that Mn(II) is the preferred cofactor for this enzyme. We herein report the unprecedented substitution of nitrosyl hydride (HNO) for dioxygen in the activity of Mn-QDO, resulting in the incorporation of both N and O atoms into the product. Turnover is demonstrated by consumption of quercetin and other related substrates under anaerobic conditions in the presence of HNO-releasing compounds and the enzyme. As with dioxygenase activity, a nonenzymatic base-catalyzed reaction of quercetin with HNO is observed above pH 7, but no enhancement of this basal reactivity is found upon addition of divalent metal salts. Unique and regioselective N-containing products ((14)N/(15)N) have been characterized by MS analysis for both the enzymatic and nonenzymatic reactions. Of the several metallo-QDO enzymes examined for nitroxygenase activity under anaerobic condition, only the Mn(II) is active; the Fe(II) and Co(II) substituted enzymes show little or no activity. This result represents an enzymatic catalysis which we denote nitroxygenase activity; the unique reactivity of the Mn-QDO suggests a metal-mediated electron transfer mechanism rather than metal activation of the substrate's inherent base-catalyzed reactivity.
Collapse
|
29
|
Benson RC, Hardy KA, Morris CR. Arginase and arginine dysregulation in asthma. J Allergy (Cairo) 2011; 2011:736319. [PMID: 21747870 PMCID: PMC3124954 DOI: 10.1155/2011/736319] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 02/07/2011] [Accepted: 02/10/2011] [Indexed: 01/01/2023] Open
Abstract
In recent years, evidence has accumulated indicating that the enzyme arginase, which converts L-arginine into L-ornithine and urea, plays a key role in the pathogenesis of pulmonary disorders such as asthma through dysregulation of L-arginine metabolism and modulation of nitric oxide (NO) homeostasis. Allergic asthma is characterized by airway hyperresponsiveness, inflammation, and remodeling. Through substrate competition, arginase decreases bioavailability of L-arginine for nitric oxide synthase (NOS), thereby limiting NO production with subsequent effects on airway tone and inflammation. By decreasing L-arginine bioavailability, arginase may also contribute to the uncoupling of NOS and the formation of the proinflammatory oxidant peroxynitrite in the airways. Finally, arginase may play a role in the development of chronic airway remodeling through formation of L-ornithine with downstream production of polyamines and L-proline, which are involved in processes of cellular proliferation and collagen deposition. Further research on modulation of arginase activity and L-arginine bioavailability may reveal promising novel therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Renée C. Benson
- Bay Area Pediatric Pulmonary Medical Corporation, Children's Hospital & Research Center Oakland, Oakland, CA 94609, USA
| | - Karen A. Hardy
- Bay Area Pediatric Pulmonary Medical Corporation, Children's Hospital & Research Center Oakland, Oakland, CA 94609, USA
| | - Claudia R. Morris
- Department of Emergency Medicine, Children's Hospital & Research Center Oakland, Oakland, CA 94609, USA
| |
Collapse
|
30
|
Schulz E, Gori T, Münzel T. Oxidative stress and endothelial dysfunction in hypertension. Hypertens Res 2011; 34:665-73. [PMID: 21512515 DOI: 10.1038/hr.2011.39] [Citation(s) in RCA: 336] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Systemic arterial hypertension is a highly prevalent cardiovascular risk factor that causes significant morbidity and mortality, and is becoming an increasingly common health problem because of the increasing longevity and prevalence of predisposing factors such as sedentary lifestyle, obesity and nutritional habits. Further complicating the impact of this disease, mild and moderate hypertension are usually asymptomatic, and their presence (and the subsequent increase in cardiovascular risk) is often unrecognized. The pathophysiology of hypertension involves a complex interaction of multiple vascular effectors including the activation of the sympathetic nervous system, of the renin-angiotensin-aldosterone system and of the inflammatory mediators. Subsequent vasoconstriction and inflammation ensue, leading to vessel wall remodeling and, finally, to the formation of atherosclerotic lesions as the hallmark of advanced disease. Oxidative stress and endothelial dysfunction are consistently observed in hypertensive subjects, but emerging evidence suggests that they also have a causal role in the molecular processes leading to hypertension. Reactive oxygen species (ROS) may directly alter vascular function or cause changes in vascular tone by several mechanisms including altered nitric oxide (NO) bioavailability or signaling. ROS-producing enzymes involved in the increased vascular oxidative stress observed during hypertension include the NADPH oxidase, xanthine oxidase, the mitochondrial respiratory chain and an uncoupled endothelial NO synthase. In the current review, we will summarize our current understanding of the molecular mechanisms in the development of hypertension with an emphasis on oxidative stress and endothelial dysfunction.
Collapse
Affiliation(s)
- Eberhard Schulz
- II. Medizinische Klinik, Universitätsmedizin Mainz, Kardiologie, Angiologie und Internistische Intensivmedizin, Mainz, Germany
| | | | | |
Collapse
|
31
|
Acute DPP-4 inhibition modulates vascular tone through GLP-1 independent pathways. Vascul Pharmacol 2011; 55:2-9. [PMID: 21397040 DOI: 10.1016/j.vph.2011.03.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/28/2011] [Accepted: 03/03/2011] [Indexed: 12/25/2022]
Abstract
Evidence from both clinical and experimental studies indicates that Di-peptidyl peptidase-IV (DPP-4) inhibition may mediate favorable effects on the cardiovascular system. The objective of this study was to examine the acute effects of DPP-4 inhibition on vascular responses and to study the underlying mechanisms of alteration in tone. Aortic segments from C57BL/6 mice were treated with vasoconstrictors and exposed to various doses of alogliptin, a selective DPP-4 inhibitor. Vasodilator responses were evaluated using pathway specific antagonists to elucidate mechanisms of response. In parallel experiments, cultured human umbilical vein endothelial cells (HUVEC) were exposed to varying concentrations of alogliptin to evaluate the effects on candidate vasodilator pathways. Alogliptin relaxed phenylephrine and U46619 pre-constricted aortic segments in a dose dependent manner. Relaxation responses were not affected by the glucagon-like peptide-1 (GLP-1) receptor antagonist, exendin fragment 9-39 (88 ± 6 vs. 91 ± 2, p < 0.001). Vascular relaxation to alogliptin was significantly decreased by endothelial denudation, L-N(G)-monomethyl-arginine citrate (L-NMMA) and by the soluble guanylate cyclase inhibitor ODQ. DPP-4 inhibition induced relaxation was completely abolished by a combination of L-NMMA, charybdotoxin and apamin. Incubation of HUVECs with alogliptin resulted in eNOS and Akt phosphorylation (Ser(1177) and Ser(473) respectively) paralleled by a rapid increase in nitric oxide. Inhibition of Src kinase decreased eNOS and Akt phosphorylation, in contrast to a lack of any effect on insulin mediated activation of the eNOS-Akt, suggesting that alogliptin mediates vasodilation through Src kinase mediated effects on eNOS-Akt. DPP-4 inhibition by alogliptin mediates rapid vascular relaxation via GLP-1 independent, Src-Akt-eNOS mediated NO release and the activation of vascular potassium channels.
Collapse
|
32
|
Álvares TS, Meirelles CM, Bhambhani YN, Paschoalin VM, Gomes PS. L-Arginine as a Potential Ergogenic Aid in Healthy Subjects. Sports Med 2011; 41:233-48. [DOI: 10.2165/11538590-000000000-00000] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
33
|
Nitrite reduction by xanthine oxidase family enzymes: a new class of nitrite reductases. J Biol Inorg Chem 2010; 16:443-60. [DOI: 10.1007/s00775-010-0741-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 11/19/2010] [Indexed: 02/04/2023]
|
34
|
Bencsik P, Kupai K, Giricz Z, Görbe A, Pipis J, Murlasits Z, Kocsis GF, Varga-Orvos Z, Puskás LG, Csonka C, Csont T, Ferdinandy P. Role of iNOS and peroxynitrite–matrix metalloproteinase-2 signaling in myocardial late preconditioning in rats. Am J Physiol Heart Circ Physiol 2010; 299:H512-8. [DOI: 10.1152/ajpheart.00052.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We have previously shown that the inhibition of myocardial nitric oxide (NO) and peroxynitrite-matrix metalloproteinase (MMP) signaling by early preconditioning (PC) is involved in its cardioprotective effect. Therefore, in the present study, we investigated the role of NO and peroxynitrite-MMP signaling in the development of late PC. PC was performed by five consecutive cycles of 4-min coronary occlusion and 4-min reperfusion in anesthetized rats in vivo. Twenty-four hours later, hearts were subjected to a 30-min coronary occlusion followed by 180-min reperfusion to measure infarct size. In separate experiments, heart tissue was sampled to measure biochemical parameters before and 3, 6, 12, or 24 h after the PC protocol, respectively. Late PC decreased infarct size, increased cardiac inducible NO synthase (iNOS) activity and gene expression, and decreased SOD activity at 24 h significantly compared with sham-operated controls. Late PC increased cardiac superoxide levels significantly at 24 h; however, it did not change cardiac NO levels. Cardiac peroxynitrite levels were significantly decreased. Downstream cellular targets of peroxynitrite, MMP-2 and MMP-9 activities were decreased in the late PC group at 24 h compared with the sham-operated group. To verify if PC-induced inhibition of MMPs had a causative role in the reduction of infarct size, in separate experiments, we measured infarct size after the pharmacological inhibition of MMPs by ilomastat and found a significant reduction of infarct size compared with the vehicle-treated group. In conclusion, this is the first demonstration that the inhibition of cardiac peroxynitrite-MMP signaling contributes to cardioprotection by late PC and that pharmacological inhibition of MMPs is able to reduce infarct size in vivo. Furthermore, increased expression of iNOS may play a role in the development of late PC; however, increased iNOS activity does not lead to increased NO production in late PC.
Collapse
Affiliation(s)
- Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Krisztina Kupai
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
| | - Zoltán Giricz
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Judit Pipis
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Zsolt Murlasits
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Gabriella F. Kocsis
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
| | - Zoltán Varga-Orvos
- Laboratory of Functional Genomics Biological Research Center, Hungarian Academy of Sciences; and
| | - László G. Puskás
- Laboratory of Functional Genomics Biological Research Center, Hungarian Academy of Sciences; and
| | - Csaba Csonka
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Tamás Csont
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
35
|
Samuni U, Samuni Y, Goldstein S. On the Distinction between Nitroxyl and Nitric Oxide Using Nitronyl Nitroxides. J Am Chem Soc 2010; 132:8428-32. [DOI: 10.1021/ja101945j] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Uri Samuni
- Department of Chemistry and Biochemistry, Queens College, City University of New York, Flushing, New York 11367, and Department of Prosthodontics, School of Dental Medicine, and Institute of Chemistry, The Accelerator Laboratory, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Yuval Samuni
- Department of Chemistry and Biochemistry, Queens College, City University of New York, Flushing, New York 11367, and Department of Prosthodontics, School of Dental Medicine, and Institute of Chemistry, The Accelerator Laboratory, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Sara Goldstein
- Department of Chemistry and Biochemistry, Queens College, City University of New York, Flushing, New York 11367, and Department of Prosthodontics, School of Dental Medicine, and Institute of Chemistry, The Accelerator Laboratory, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
36
|
Zweier JL, Li H, Samouilov A, Liu X. Mechanisms of nitrite reduction to nitric oxide in the heart and vessel wall. Nitric Oxide 2010; 22:83-90. [PMID: 20044016 DOI: 10.1016/j.niox.2009.12.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 12/10/2009] [Accepted: 12/21/2009] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is an important regulator of a variety of biological functions, and also has a role in the pathogenesis of cellular injury. It had been generally accepted that NO is solely generated in biological tissues by specific nitric oxide synthases (NOS) which metabolize arginine to citrulline with the formation of NO. However, over the last 15 years, nitrite-mediated NO production has been shown to be an important mechanism of NO formation in the heart and cardiovascular system. Now numerous studies have demonstrated that nitrite can be an important source rather than simply a product of NO in mammalian cells and tissues and can be a potential vasodilator drug for cardiovascular diseases. There are a variety of mechanisms of nitrite reduction to NO and it is now appreciated that this process, while enhanced under hypoxic conditions, also occurs under normoxia. Several methods, including electron paramagnetic resonance, chemiluminescence NO analyzer, and NO electrode have been utilized to measure, quantitate, and image nitrite-mediated NO formation. Results reveal that nitrite-dependent NO generation plays critical physiological and pathological roles, and is controlled by oxygen tension, pH, reducing substrates and nitrite levels. In this manuscript, we review the mechanisms of nitrite-mediated NO formation and the effects of oxygen on this process with a focus on how this occurs in the heart and vessels.
Collapse
Affiliation(s)
- Jay L Zweier
- Davis Heart and Lung Research Institute, The Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, 473 West 12th Avenue, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
37
|
Chen K, Pittman RN, Popel AS. Hemorrhagic shock and nitric oxide release from erythrocytic nitric oxide synthase: a quantitative analysis. Microvasc Res 2009; 78:107-18. [PMID: 19285090 PMCID: PMC2782400 DOI: 10.1016/j.mvr.2009.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 02/21/2009] [Accepted: 02/23/2009] [Indexed: 01/15/2023]
Abstract
A large loss of blood during hemorrhage can result in profound shock, a state of hypotension associated with hemodynamic abnormalities. One of the hypotheses to account for this collapse of homeostasis is that the production of nitric oxide (NO), a gas molecule that dilates blood vessels, is significantly impaired during hemorrhage, resulting in a mismatch between O(2) delivery and the metabolic activity in the tissues. NO can be released from multiple sources in the vasculature. Recent studies have shown that erythrocytes express functional endothelial nitric oxide synthase (NOS3), which potentially serves as an intraluminal NO source. NO delivery from this source is complex: erythrocytes are not only NO producers but also act as potent sinks because of the high affinity of NO for hemoglobin. To test our hypothesis that the loss of erythrocytic NOS3 during hemorrhage contributes to NO deficiency-related shock, we have constructed a multicellular computational model that simulates NO production and transport to allow us to quantify the loss of NO under different hemorrhagic conditions. Our model shows that: (1) during mild hemorrhage and subsequent hemodilution (hematocrit >30%), NO from this intraluminal source is only slightly decreased in the vascular smooth muscle, but the NO level is significantly reduced under severe hemorrhagic conditions (hematocrit <30%); (2) whether a significant amount of NO from this source can be delivered to vascular smooth muscle is strongly dependent on the existence of a protective mechanism for NO delivery; (3) if the expression level of NOS3 on erythrocytes is similar to that on endothelial cells, we estimate approximately 13 pM NO at the vascular smooth muscle from this source when such a protective mechanism is involved. This study provides a basis for detailed studies to characterize the impairment of NO release pathways during hemorrhage and yield important insights for the development of resuscitation methods.
Collapse
Affiliation(s)
- Kejing Chen
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 613 Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
38
|
Continuous electrochemical monitoring of nitric oxide production in murine macrophage cell line RAW 264.7. Anal Bioanal Chem 2009; 394:1497-504. [DOI: 10.1007/s00216-009-2813-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 04/17/2009] [Accepted: 04/17/2009] [Indexed: 02/07/2023]
|
39
|
Gur S, Kadowitz PJ, Hellstrom WJG. A critical appraisal of erectile function in animal models of diabetes mellitus. ACTA ACUST UNITED AC 2009; 32:93-114. [DOI: 10.1111/j.1365-2605.2008.00928.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
40
|
McCarthy HO, Coulter JA, Robson T, Hirst DG. Gene therapy via inducible nitric oxide synthase: a tool for the treatment of a diverse range of pathological conditions. J Pharm Pharmacol 2008; 60:999-1017. [PMID: 18644193 DOI: 10.1211/jpp.60.8.0007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO(.)) is a reactive nitrogen radical produced by the NO synthase (NOS) enzymes; it affects a plethora of downstream physiological and pathological processes. The past two decades have seen an explosion in the understanding of the role of NO(.) biology, highlighting various protective and damaging modes of action. Much of the controversy surrounding the role of NO(.) relates to the differing concentrations generated by the three isoforms of NOS. Both calcium-dependent isoforms of the enzyme (endothelial and neuronal NOS) generate low-nanomolar/picomolar concentrations of NO(.). By contrast, the calcium-independent isoform (inducible NOS (iNOS)) generates high concentrations of NO(.), 2-3 orders of magnitude greater. This review summarizes the current literature in relation to iNOS gene therapy for the therapeutic benefit of various pathological conditions, including various states of vascular disease, wound healing, erectile dysfunction, renal dysfunction and oncology. The available data provide convincing evidence that manipulation of endogenous NO(.) using iNOS gene therapy can provide the basis for future clinical trials.
Collapse
Affiliation(s)
- Helen O McCarthy
- School of Pharmacy, McClay Research Centre, Queen's University, Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | | | | | | |
Collapse
|
41
|
Hu Z, Chen J, Wei Q, Xia Y. Bidirectional actions of hydrogen peroxide on endothelial nitric-oxide synthase phosphorylation and function: co-commitment and interplay of Akt and AMPK. J Biol Chem 2008; 283:25256-25263. [PMID: 18617528 DOI: 10.1074/jbc.m802455200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Endothelial NO synthase (eNOS) is critically modulated by kinases via the phosphorylation of its Ser(1179) (bovine) or Ser(1177) (human) residue. Reactive oxygen species such as H(2)O(2) was reported to activate Akt, leading to increased eNOS Ser(1179) phosphorylation and activity. But reactive oxygen species are also known to attenuate eNOS function in cardiovascular diseases. Prior studies showing H(2)O(2)-stimulated eNOS phosphorylation were performed on serum-starved cells, and only the short term effect of H(2)O(2) was examined. Here we found that the effects of H(2)O(2) on eNOS Ser(1179) phosphorylation and function were bidirectional. With endothelial cells cultured with serum, H(2)O(2) initially raised eNOS Ser(1179) phosphorylation and activity. However, after the peak increase at 30 min, eNOS Ser(1179) phosphorylation dramatically declined. Parallel to the alterations of eNOS Ser(1179) phosphorylation, Akt was transiently activated by H(2)O(2) and subsequently became dormant. In contrast, AMP-activated protein kinase (AMPK) was progressively activated in H(2)O(2)-treated cells. Blocking Akt activation abolished the initial rise of eNOS Ser(1179) phosphorylation after H(2)O(2) treatment. In long term H(2)O(2)-treated cells where Akt was deactivated, significant amounts of Ser(1179)-phosphorylated eNOS remained. AMPK inhibition eradicated the remaining eNOS Ser(1179) phosphorylation. Taken together, these studies revealed that Akt and AMPK orchestrated a bidirectional action on eNOS Ser(1179) phosphorylation in H(2)O(2)-treated cells. Long term H(2)O(2) exposure decreased eNOS Ser(1179) phosphorylation, and this might account for the loss of eNOS function in cardiovascular diseases where chronic oxidative injury occurs.
Collapse
Affiliation(s)
- Zhuangli Hu
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Molecular and Cellular Biochemistry, The Ohio State University Medical Center, Columbus, Ohio 43210
| | - Juan Chen
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Molecular and Cellular Biochemistry, The Ohio State University Medical Center, Columbus, Ohio 43210
| | - Qin Wei
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Molecular and Cellular Biochemistry, The Ohio State University Medical Center, Columbus, Ohio 43210
| | - Yong Xia
- Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, Department of Molecular and Cellular Biochemistry, The Ohio State University Medical Center, Columbus, Ohio 43210.
| |
Collapse
|
42
|
Schulz E, Jansen T, Wenzel P, Daiber A, Münzel T. Nitric oxide, tetrahydrobiopterin, oxidative stress, and endothelial dysfunction in hypertension. Antioxid Redox Signal 2008; 10:1115-26. [PMID: 18321209 DOI: 10.1089/ars.2007.1989] [Citation(s) in RCA: 320] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial dysfunction in the setting of cardiovascular risk factors such as hypercholesterolemia, diabetes mellitus, chronic smoking, as well hypertension, is, at least in part, dependent of the production of reactive oxygen species (ROS) and the subsequent decrease in vascular bioavailability of nitric oxide (NO). ROS-producing enzymes involved in increased oxidative stress within vascular tissue include NADPH oxidase, xanthine oxidase, and mitochondrial superoxide producing enzymes. Superoxide produced by the NADPH oxidase may react with NO, thereby stimulating the production of the NO/superoxide reaction product peroxynitrite. Peroxynitrite in turn has been shown to uncouple eNOS, therefore switching an antiatherosclerotic NO producing enzyme to an enzyme that may accelerate the atherosclerotic process by producing superoxide. Increased oxidative stress in the vasculature, however, is not restricted to the endothelium and also occurs within the smooth muscle cell layer. Increased superoxide production has important consequences with respect to signaling by the soluble guanylate cyclase and the cGMP-dependent kinase I, which activity and expression is regulated in a redox-sensitive fashion. The present review will summarize current concepts concerning eNOS uncoupling, with special focus on the role of tetrahydrobiopterin in mediating eNOS uncoupling.
Collapse
Affiliation(s)
- Eberhard Schulz
- II Medizinische Klinik, Mainz, Kardiologie, Angiologie und Internistische Intensivmedizin, Mainz, Germany
| | | | | | | | | |
Collapse
|
43
|
Wheatley C. The return of the Scarlet Pimpernel: cobalamin in inflammation II - cobalamins can both selectively promote all three nitric oxide synthases (NOS), particularly iNOS and eNOS, and, as needed, selectively inhibit iNOS and nNOS. JOURNAL OF NUTRITIONAL & ENVIRONMENTAL MEDICINE 2007; 16:181-211. [PMID: 18836533 PMCID: PMC2556189 DOI: 10.1080/10520290701791839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The up-regulation of transcobalamins [hitherto posited as indicating a central need for cobalamin (Cbl) in inflammation], whose expression, like inducible nitric oxide synthase (iNOS), is Sp1- and interferondependent, together with increased intracellular formation of glutathionylcobalamin (GSCbl), adenosylcobalamin (AdoCbl), methylcobalamin (MeCbl), may be essential for the timely promotion and later selective inhibition of iNOS and concordant regulation of endothelial and neuronal NOS (eNOS/nNOS.) Cbl may ensure controlled high output of nitric oxide (NO) and its safe deployment, because: (1) Cbl is ultimately responsible for the synthesis or availability of the NOS substrates and cofactors heme, arginine, BH(4) flavin adenine dinucleotide/flavin mononucleotide (FAD/FMN) and NADPH, via the far-reaching effects of the two Cbl coenzymes, methionine synthase (MS) and methylmalonyl CoA mutase (MCoAM) in, or on, the folate, glutathione, tricarboxylic acid (TCA) and urea cycles, oxidative phosphorylation, glycolysis and the pentose phosphate pathway. Deficiency of any of theNOS substrates and cofactors results in 'uncoupled' NOS reactions, decreasedNO production and increased or excessive O(2) (-), H(2)O(2), ONOO(-) and other reactive oxygen species (ROS), reactive nitric oxide species (RNIS) leading to pathology. (2) Cbl is also the overlooked ultimate determinant of positive glutathione status, which favours the formation of more benign NO species, s-nitrosothiols, the predominant form in which NO is safely deployed. Cbl status may consequently act as a 'back-up disc' that ensures the active status of antioxidant systems, as well as reversing and modulating the effects of nitrosylation in cell signal transduction.New evidence shows that GSCbl can significantly promote iNOS/ eNOS NO synthesis in the early stages of inflammation, thus lowering high levels of tumour necrosis factor-a that normally result in pathology, while existing evidence shows that in extreme nitrosative and oxidative stress, GSCbl can regenerate the activity of enzymes important for eventual resolution, such as glucose 6 phosphate dehydrogenase, which ensures NADPH supply, lactate dehydrogenase, and more; with human clinical case studies of OHCbl for cyanide poisoning, suggesting Cbl may regenerate aconitase and cytochrome c oxidase in the TCA cycle and oxidative phosphorylation. Thus, Cbl may simultaneously promote a strong inflammatory response and the means to resolve it.
Collapse
Affiliation(s)
- Carmen Wheatley
- Orthomolecular Oncology, 4 Richmond Road, Oxford OX1 2JJ, UK
| |
Collapse
|
44
|
Podesser BK, Hallström S. Nitric oxide homeostasis as a target for drug additives to cardioplegia. Br J Pharmacol 2007; 151:930-40. [PMID: 17486142 PMCID: PMC2042932 DOI: 10.1038/sj.bjp.0707272] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2006] [Revised: 03/06/2007] [Accepted: 04/02/2007] [Indexed: 11/09/2022] Open
Abstract
The vascular endothelium of the coronary arteries has been identified as the important organ that locally regulates coronary perfusion and cardiac function by paracrine secretion of nitric oxide (NO) and vasoactive peptides. NO is constitutively produced in endothelial cells by endothelial nitric oxide synthase (eNOS). NO derived from this enzyme exerts important biological functions including vasodilatation, scavenging of superoxide and inhibition of platelet aggregation. Routine cardiac surgery or cardiologic interventions lead to a serious temporary or persistent disturbance in NO homeostasis. The clinical consequences are "endothelial dysfunction", leading to "myocardial dysfunction": no- or low-reflow phenomenon and temporary reduction of myocardial pump function. Uncoupling of eNOS (one electron transfer to molecular oxygen, the second substrate of eNOS) during ischemia-reperfusion due to diminished availability of L-arginine and/or tetrahydrobiopterin is even discussed as one major source of superoxide formation. Therefore maintenance of normal NO homeostasis seems to be an important factor protecting from ischemia/reperfusion (I/R) injury. Both, the clinical situations of cardioplegic arrest as well as hypothermic cardioplegic storage are followed by reperfusion. However, the presently used cardioplegic solutions to arrest and/or store the heart, thereby reducing myocardial oxygen consumption and metabolism, are designed to preserve myocytes mainly and not endothelial cells. This review will focus on possible drug additives to cardioplegia, which may help to maintain normal NO homeostasis after I/R.
Collapse
Affiliation(s)
- B K Podesser
- The Ludwig Boltzmann Cluster for Cardiovascular Research, Medical University of Vienna Vienna, Austria
| | - S Hallström
- Institute of Physiological Chemistry, Center for Physiological Medicine, Medical University of Graz Graz, Austria
| |
Collapse
|
45
|
Sáenz DA, Bari SE, Salido E, Chianelli M, Rosenstein RE. Effect of nitroxyl on the hamster retinal nitridergic pathway. Neurochem Int 2007; 51:424-32. [PMID: 17543420 DOI: 10.1016/j.neuint.2007.04.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2006] [Revised: 03/30/2007] [Accepted: 04/23/2007] [Indexed: 10/23/2022]
Abstract
There is a growing body of evidence on the role of nitric oxide (NO) in retinal physiology. Recently, interest has developed in the functional role of an alternative redox form of NO, namely nitroxyl (HNO/NO(-)), because it is formed by a number of diverse biochemical reactions. The aim of the present report was to comparatively analyze the effect of HNO and NO on the retinal nitridergic pathway in the golden hamster. For this purpose, sodium trioxodinitrate (Angeli's salt) and diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA/NO) were used as HNO and NO releasers, respectively. Angeli's salt and DEA/NO significantly decreased nitric oxide synthase activity. In addition, Angeli's salt (but not DEA/NO) significantly decreased l-arginine uptake. DEA/NO significantly increased cGMP accumulation at low micromolar concentrations, while Angeli's salt affected this parameter with a threshold concentration of 200muM. Although Angeli's salt and DEA/NO significantly diminished reduced glutathione and protein thiol levels in a similar way, DEA/NO was significantly more effective than AS in increasing S-nitrosothiol levels. None of these compounds increased retinal lipid peroxidation. These results suggest that HNO could regulate the hamster retinal nitridergic pathway by acting through a mechanism that only partly overlaps with that involved in NO response.
Collapse
Affiliation(s)
- Daniel A Sáenz
- Laboratorio de Neuroquímica Retiniana y Oftalmología Experimental, Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, CEFyBO, CONICET, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
46
|
Zhang P, Xu X, Hu X, van Deel ED, Zhu G, Chen Y. Inducible nitric oxide synthase deficiency protects the heart from systolic overload-induced ventricular hypertrophy and congestive heart failure. Circ Res 2007; 100:1089-98. [PMID: 17363700 PMCID: PMC2386857 DOI: 10.1161/01.res.0000264081.78659.45] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inducible nitric oxide synthase (iNOS) protein is expressed in cardiac myocytes of patients and experimental animals with congestive heart failure (CHF). Here we show that iNOS expression plays a role in pressure overload-induced myocardial chamber dilation and hypertrophy. In wild-type mice, chronic transverse aortic constriction (TAC) resulted in myocardial iNOS expression, cardiac hypertrophy, ventricular dilation and dysfunction, and fibrosis, whereas iNOS-deficient mice displayed much less hypertrophy, dilation, fibrosis, and dysfunction. Consistent with these findings, TAC resulted in marked increases of myocardial atrial natriuretic peptide 4-hydroxy-2-nonenal (a marker of lipid peroxidation) and nitrotyrosine (a marker for peroxynitrite) in wild-type mice but not in iNOS-deficient mice. In response to TAC, myocardial endothelial NO synthase and iNOS was expressed as both monomer and dimer in wild-type mice, and this was associated with increased reactive oxygen species production, suggesting that iNOS monomer was a source for the increased oxidative stress. Moreover, systolic overload-induced Akt, mammalian target of rapamycin, and ribosomal protein S6 activation was significantly attenuated in iNOS-deficient mice. Furthermore, selective iNOS inhibition with 1400W (6 mg/kg per hour) significantly attenuated TAC induced myocardial hypertrophy and pulmonary congestion. These data implicate iNOS in the maladaptative response to systolic overload and suggest that selective iNOS inhibition or attenuation of iNOS monomer content might be effective for treatment of systolic overload-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Ping Zhang
- Center of Vascular Biology and Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
47
|
Bae SR, Wu GS, Sevanian A, Schultz BE, Zamir E, Rao NA. Direct detection of reactive nitrogen species in experimental autoimmune uveitis. KOREAN JOURNAL OF OPHTHALMOLOGY 2007; 21:21-7. [PMID: 17460428 PMCID: PMC2629686 DOI: 10.3341/kjo.2007.21.1.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 02/02/2007] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Demonstrate unequivocally the generation of nitric oxide in experimental autoimmune uveoretinitis by electron spin resonance spectroscopy (ESR) using ferrous iron complex of N-methyl-D-glucamine dithiocarbamate, (MGD)(2)-Fe(2+), as a spin trap. METHODS Experimental autoimmune uveitis was induced in Lewis rats, and at the peak of the intraocular inflammation, the animals received intravitreous injections of the spin trap. The retina and choroid dissected from the enucleated globes were subjected to ESR. Similarly, the retina and choroid obtained at the peak of experimental autoimmune uveo-retinitis (EAU) were placed in a vial containing luminal, and chemiluminescence was counted on a Packard liquid scintillation analyzer. RESULTS The ESR three-line spectrum (g=2.04; a(N)=12.5 G) obtained was characteristic of the adduct [(MGD)(2)-Fe(2+)-NO]. The majority of this signal was eliminated by the inducible nitric oxide synthase (iNOS) specific inhibitor aminoguanidine injected inflamed retina was detected when compared with that of the non inflamed controls. The chemiluminescent activity was further increased two-fold by the addition of bicarbonate to the inflamed retina; the phenomenon is attributable only to the presence of a high steady-state concentration of peroxynitrite. CONCLUSIONS The study shows an unequivocal presence of nitric oxide in EAU retina and choroid and the generation of peroxynitrite. High levels of these reactive nitrogen species generated in the inflamed retina and choroids are certain to cause irreversible tissue damage, especially at the susceptible sites such as photoreceptors.
Collapse
Affiliation(s)
- Sun Ryang Bae
- Department of Ophthalmology & Laboratory of Visual Science, College of Medicine, The Catholic University of Korea, Daejon St. Marys Hospital, Jung-gu, Daejon, Korea.
| | | | | | | | | | | |
Collapse
|
48
|
Samouilov A, Woldman YY, Zweier JL, Khramtsov VV. Magnetic resonance study of the transmembrane nitrite diffusion. Nitric Oxide 2007; 16:362-70. [PMID: 17306575 PMCID: PMC2709508 DOI: 10.1016/j.niox.2006.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 12/15/2006] [Accepted: 12/23/2006] [Indexed: 01/08/2023]
Abstract
Nitrite (NO(2)-), being a product of metabolism of both nitric oxide (NO(*)) and nitrate (NO(3)-), can accumulate in tissues and regenerate NO() by several mechanisms. The effect of NO(2)- on ischemia/reperfusion injury was also reported. Nevertheless, the mechanisms of intracellular NO(2)- accumulation are poorly understood. We suggested significant role of nitrite penetration through biological membranes in the form of undissociated nitrous acid (HNO(2)). This hypothesis has been tested using large unilamellar phosphatidylcholine liposomes and several spectroscopic techniques. HNO(2) transport across the phospholipid bilayer of liposomes facilitates proton transfer resulting in intraliposomal acidification, which was measured using pH-sensitive probes. NO(2)(-)-mediated intraliposomal acidification was confirmed by EPR spectroscopy using membrane-impermeable pH-sensitive nitroxide, AMC (2,2,5,5-tetramethyl-1-yloxy-2,5-dihydro-1H-imidazol-3-ium-4-yl)-aminomethanesulfonic acid (pK 5.25), and by (31)P NMR spectroscopy using inorganic phosphate (pK 6.9). Nitrite accumulates inside liposomes in concentration exceeding its concentration in the bulk solution, when initial transmembrane pH gradient (alkaline inside) is applied. Intraliposomal accumulation of NO(2)- was observed by direct measurement using chemiluminescence technique. Perfusion of isolated rat hearts with buffer containing 4 microM NO(2)- was performed. The nitrite concentrations in the effluent and in the tissue, measured after 1 min perfusion, were close, supporting fast penetration of the nitrite through the tissue. Measurements of the nitrite/nitrate showed that total concentration of NO(x) in myocardium increased from initial 7.8 to 24.7 microM after nitrite perfusion. Physiological significance of passive transmembrane transport of NO(2)- and its coupling with intraliposomal acidification are discussed.
Collapse
Affiliation(s)
- A Samouilov
- Center for Biomedical EPR Spectroscopy and Imaging, The Davis Heart and Lung Research Institute, The Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
49
|
van Faassen E, Vanin A. NO trapping in biological systems with a functionalized zeolite network. Nitric Oxide 2006; 15:233-40. [PMID: 16458549 DOI: 10.1016/j.niox.2005.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 11/23/2005] [Accepted: 12/11/2005] [Indexed: 11/25/2022]
Abstract
Zeolite-Y powder has been functionalized with ferric iron-diethyldithiocarbamate complexes and applied to trap nitric oxide radicals in liquids and biological systems. The complexes have been assembled in situ in the pores of zeolite-Y and act as traps for nitric oxide radicals. The resulting mononitrosyl-iron complexes form a mixture of diamagnetic ferric and paramagnetic ferrous complexes. The yield of trapped NO may be determined ex situ using electron paramagnetic resonance. The material may be anchored on solid surfaces, mixed into a composite or compressed into small pellets. The material was used to detect endogenous NO in endothelial cell cultures and spinach leaves. The sensitivity of the functionalized zeolite is significantly better than that achieved in conventional trapping of NO with iron-diethyldithiocarbamate complexes.
Collapse
Affiliation(s)
- E van Faassen
- Interface Physics, Debye Institute, Utrecht University, Princetonplein 1, 3508 TA Utrecht, The Netherlands.
| | | |
Collapse
|
50
|
Varol Tas F, Guvenir T, Tas G, Cakaloz B, Ormen M. Nitric oxide levels in disruptive behavioral disorder. Neuropsychobiology 2006; 53:176-80. [PMID: 16763377 DOI: 10.1159/000093781] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2005] [Accepted: 03/16/2006] [Indexed: 11/19/2022]
Abstract
There are various evidences of the role of nitric oxide (NO) in several neuropsychiatric disorders. However, there is no clinical study which investigated the role of NO in disruptive behavioral disorders (DBD). The aim of this study is to investigate the relation between NO levels and DBD. NO levels were measured in serum from 45 patients diagnosed as having DBD (30 patients with a diagnosis of attention deficit and hyperactivity disorder [ADHD] and 15 with ADHD + oppositional defiant disorder [ODD]) and 51 healthy control subjects. It is statistically significant that the pure ADHD group's blood NO levels are lower than those of both the ADHD + ODD and control groups. There was no significant difference between the ADHD + ODD group and the controls. The difference of the NO levels in DBD may indicate the effect of NO in the etiology of this disorder spectrum.
Collapse
Affiliation(s)
- Fatma Varol Tas
- Department of Child and Adolescent Psychiatry, Faculty of Medicine, University of Dokuz Eylul, Izmir, Turkey.
| | | | | | | | | |
Collapse
|