1
|
Liao CJ, Tseng YT, Cheng YA, Dayao LA, Iffland-Mühlhaus L, Gee LB, Ribson RD, Chan TS, Apfel UP, Lu TT. Ligand Control of Dinitrosyl Iron Complexes for Selective Superoxide-Mediated Nitric Oxide Monooxygenation and Superoxide-Dioxygen Interconversion. J Am Chem Soc 2023; 145:20389-20402. [PMID: 37683125 DOI: 10.1021/jacs.3c05577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Through nitrosylation of [Fe-S] proteins, or the chelatable iron pool, a dinitrosyl iron unit (DNIU) [Fe(NO)2] embedded in the form of low-molecular-weight/protein-bound dinitrosyl iron complexes (DNICs) was discovered as a metallocofactor assembled under inflammatory conditions with elevated levels of nitric oxide (NO) and superoxide (O2-). In an attempt to gain biomimetic insights into the unexplored transformations of the DNIU under inflammation, we investigated the reactivity toward O2- by a series of DNICs [(NO)2Fe(μ-MePyr)2Fe(NO)2] (1) and [(NO)2Fe(μ-SEt)2Fe(NO)2] (3). During the superoxide-induced conversion of DNIC 1 into DNIC [(K-18-crown-6-ether)2(NO2)][Fe(μ-MePyr)4(μ-O)2(Fe(NO)2)4] (2-K-crown) and a [Fe3+(MePyr)x(NO2)y(O)z]n adduct, stoichiometric NO monooxygenation yielding NO2- occurs without the transient formation of peroxynitrite-derived •OH/•NO2 species. To study the isoelectronic reaction of O2(g) and one-electron-reduced DNIC 1, a DNIC featuring an electronically localized {Fe(NO)2}9-{Fe(NO)2}10 electronic structure, [K-18-crown-6-ether][(NO)2Fe(μ-MePyr)2Fe(NO)2] (1-red), was successfully synthesized and characterized. Oxygenation of DNIC 1-red leads to the similar assembly of DNIC 2-K-crown, of which the electronic structure is best described as paramagnetic with weak antiferromagnetic coupling among the four S = 1/2 {FeIII(NO-)2}9 units and S = 5/2 Fe3+ center. In contrast to DNICs 1 and 1-red, DNICs 3 and [K-18-crown-6-ether][(NO)2Fe(μ-SEt)2Fe(NO)2] (3-red) display a reversible equilibrium of "3 + O2- ⇋ 3-red + O2(g)", which is ascribed to the covalent [Fe(μ-SEt)2Fe] core and redox-active [Fe(NO)2] unit. Based on this study, the supporting/bridging ligands in dinuclear DNIC 1/3 (or 1-red/3-red) control the selective monooxygenation of NO and redox interconversion between O2- and O2 during reaction with O2- (or O2).
Collapse
Affiliation(s)
- Cheng-Jhe Liao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Ting Tseng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-An Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Loise Ann Dayao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Linda Iffland-Mühlhaus
- Department of Chemistry and Biochemistry, Inorganic Chemistry I, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Leland B Gee
- LCLS, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Ryan D Ribson
- LCLS, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Ting-Shan Chan
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Ulf-Peter Apfel
- Department of Chemistry and Biochemistry, Inorganic Chemistry I, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Electrosynthesis, Fraunhofer UMSICHT, 46047 Oberhausen, Germany
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
2
|
Vanin AF, Telegina VI, Mikoyan VD, Tkachev NA, Vasilieva SV. The Cytostatic Action of Dinitrosyl Iron Complexes with Glutathione on Escherichia coli Cells Is Mediated by Nitrosonium Cations Released from These Complexes. Biophysics (Nagoya-shi) 2022; 67:761-767. [PMID: 36567970 PMCID: PMC9762666 DOI: 10.1134/s0006350922050207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/23/2022] Open
Abstract
This study demonstrates a bacteriostatic effect of binuclear dinitrosyl iron complexes with glutathione on Escherichia coli TN300 cells. It has been quantified by the colony formation assay. The bacteriostatic effect exerted by these complexes increases considerably in the presence of diethyldithiocarbamate. Our results suggest that this effect is caused by the intense release of nitrosonium cations, NO+, from the complexes, which decompose under the action of diethyldithiocarbamate. A similar effect is observed when E. coli cells are treated with diethyldithiocarbamate 40 min after the addition of sodium nitrite or S-nitrosoglutathione. Notably, the level of dinitrosyl iron complexes observed in the bacterial cells due to the effects of sodium nitrite or S-nitrosoglutathione is almost the same as that obtained after treatment with glutathione-containing complexes. The bacteriostatic effects of the NO molecules released from nitrite or S-nitrosoglutathione during their brief interaction with bacteria were significantly smaller than the bacteriostatic effect of NO+. We deduce therefrom that the nitrosonium cations released from DNICs are responsible for the observed bacteriostatic effect of these complexes in E. coli cells.
Collapse
Affiliation(s)
- A. F. Vanin
- Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - V. I. Telegina
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - V. D. Mikoyan
- Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - N. A. Tkachev
- Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| | - S. V. Vasilieva
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
3
|
The Relationship of Glutathione- S-Transferase and Multi-Drug Resistance-Related Protein 1 in Nitric Oxide (NO) Transport and Storage. Molecules 2021; 26:molecules26195784. [PMID: 34641326 PMCID: PMC8510172 DOI: 10.3390/molecules26195784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
Nitric oxide is a diatomic gas that has traditionally been viewed, particularly in the context of chemical fields, as a toxic, pungent gas that is the product of ammonia oxidation. However, nitric oxide has been associated with many biological roles including cell signaling, macrophage cytotoxicity, and vasodilation. More recently, a model for nitric oxide trafficking has been proposed where nitric oxide is regulated in the form of dinitrosyl-dithiol-iron-complexes, which are much less toxic and have a significantly greater half-life than free nitric oxide. Our laboratory has previously examined this hypothesis in tumor cells and has demonstrated that dinitrosyl-dithiol-iron-complexes are transported and stored by multi-drug resistance-related protein 1 and glutathione-S-transferase P1. A crystal structure of a dinitrosyl-dithiol-iron complex with glutathione-S-transferase P1 has been solved that demonstrates that a tyrosine residue in glutathione-S-transferase P1 is responsible for binding dinitrosyl-dithiol-iron-complexes. Considering the roles of nitric oxide in vasodilation and many other processes, a physiological model of nitric oxide transport and storage would be valuable in understanding nitric oxide physiology and pathophysiology.
Collapse
|
4
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
5
|
Palmieri EM, McGinity C, Wink DA, McVicar DW. Nitric Oxide in Macrophage Immunometabolism: Hiding in Plain Sight. Metabolites 2020; 10:metabo10110429. [PMID: 33114647 PMCID: PMC7693038 DOI: 10.3390/metabo10110429] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric Oxide (NO) is a soluble endogenous gas with various biological functions like signaling, and working as an effector molecule or metabolic regulator. In response to inflammatory signals, immune myeloid cells, like macrophages, increase production of cytokines and NO, which is important for pathogen killing. Under these proinflammatory circumstances, called “M1”, macrophages undergo a series of metabolic changes including rewiring of their tricarboxylic acid (TCA) cycle. Here, we review findings indicating that NO, through its interaction with heme and non-heme metal containing proteins, together with components of the electron transport chain, functions not only as a regulator of cell respiration, but also a modulator of intracellular cell metabolism. Moreover, diverse effects of NO and NO-derived reactive nitrogen species (RNS) involve precise interactions with different targets depending on concentration, temporal, and spatial restrictions. Although the role of NO in macrophage reprogramming has been in evidence for some time, current models have largely minimized its importance. It has, therefore, been hiding in plain sight. A review of the chemical properties of NO, past biochemical studies, and recent publications, necessitates that mechanisms of macrophage TCA reprogramming during stimulation must be re-imagined and re-interpreted as mechanistic results of NO exposure. The revised model of metabolic rewiring we describe here incorporates many early findings regarding NO biochemistry and brings NO out of hiding and to the forefront of macrophages immunometabolism.
Collapse
|
6
|
Vanin AF. How is Nitric Oxide (NO) Converted into Nitrosonium Cations (NO +) in Living Organisms? (Based on the Results of Optical and EPR Analyses of Dinitrosyl Iron Complexes with Thiol-Containing Ligands). APPLIED MAGNETIC RESONANCE 2020; 51:851-876. [PMID: 33100585 PMCID: PMC7572240 DOI: 10.1007/s00723-020-01270-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The present work provides theoretical and experimental foundations for the ability of dinitrosyl iron complexes (DNICs) with thiol-containing ligands to be not only the donors of neutral NO molecules, but also the donors of nitrosonium cations (NO+) in living organisms ensuring S-nitrosation of various proteins and low-molecular-weight compounds. It is proposed that the emergence of those cations in DNICs is related to disproportionation reaction of NO molecules, initiated by their binding with Fe2+ ions (two NO molecules per one ion). At the same time, possible hydrolysis of iron-bound nitrosonium cations is prevented by the electron density transition to nitrosonium cations from sulfur atoms of thiol-containing ligands, which are included in the coordination sphere of iron. It allows supposing that iron in iron-nitrosyl complexes of DNICs has a d 7 electronic configuration. This supposition is underpinned by experimental data revealing that a half of nitrosyl ligands are converted into S-nitrosothiols (RSNOs) when those complexes decompose, with the other half of those ligands released in the form of neutral NO molecules.
Collapse
Affiliation(s)
- Anatoly F. Vanin
- Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| |
Collapse
|
7
|
Vanin AF. The Free-Radical Nature of Nitric Oxide Molecules as a Determinant of their Conversion to Nitrosonium Cations in Living Systems. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920030239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
8
|
Vanin AF. Dinitrosyl iron complexes with thiol-containing ligands as a base for developing drugs with diverse therapeutic activities: Physicochemical and biological substantiation. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s0006350917040224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
9
|
Mikoyan VD, Burgova EN, Borodulin RR, Vanin AF. The binuclear form of dinitrosyl iron complexes with thiol-containing ligands in animal tissues. Nitric Oxide 2016; 62:1-10. [PMID: 27989818 DOI: 10.1016/j.niox.2016.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/19/2016] [Accepted: 10/27/2016] [Indexed: 11/18/2022]
Abstract
It has been established that treatment of mice with sodium nitrite, S-nitrosoglutathione and the water-soluble nitroglycerine derivative isosorbide dinitrate (ISDN) as NO donors initiates in vivo synthesis of significant amounts of EPR-silent binuclear dinitrosyl iron complexes (B-DNIC) with thiol-containing ligands in the liver and other tissues of experimental mice. This effect is especially apparent if NO donors are administered to mice simultaneously with the Fe2+-citrate complex. Similar results were obtained in experiments on isolated liver and other mouse tissues treated with gaseous NО in vitro and during stimulation of endogenous NO synthesis in the presence of inducible NO synthase. B-DNIC appeared in mouse tissues after in vitro treatment of tissue samples with an aqueous solution of diethyldithiocarbamate (DETC), which resulted in the transfer of iron-mononitrosyl fragments from B-DNIC to the thiocarbonyl group of DETC and the formation of EPR-detectable mononitrosyl iron complexes (MNIC) with DETC. EPR-Active MNIC with N-methyl-d-glucamine dithiocarbamate (MGD) were synthesized in a similar way. MNIC-MGD were also formed in the reaction of water-soluble MGD-Fe2+ complexes with sodium nitrite, S-nitrosoglutathione and ISDN.
Collapse
Affiliation(s)
- Vasak D Mikoyan
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Evgeniya N Burgova
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Rostislav R Borodulin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly F Vanin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Institute for Regenerative Medicine, I.M.Sechenov Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
10
|
Dinitrosyl iron complexes with thiol-containing ligands as a “working form” of endogenous nitric oxide. Nitric Oxide 2016; 54:15-29. [DOI: 10.1016/j.niox.2016.01.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/18/2016] [Accepted: 01/21/2016] [Indexed: 02/03/2023]
|
11
|
Vanin AF, Mikoyan VD, Kubrina LN, Borodulin RR, Burgova EN. Mono- and binuclear dinitrosyl iron complexes with thiol-containing ligands in various biosystems. Biophysics (Nagoya-shi) 2015. [DOI: 10.1134/s0006350915040247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
12
|
Lok HC, Sahni S, Richardson V, Kalinowski DS, Kovacevic Z, Lane DJR, Richardson DR. Glutathione S-transferase and MRP1 form an integrated system involved in the storage and transport of dinitrosyl-dithiolato iron complexes in cells. Free Radic Biol Med 2014; 75:14-29. [PMID: 25035074 DOI: 10.1016/j.freeradbiomed.2014.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/26/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022]
Abstract
Nitrogen monoxide (NO) is vital for many essential biological processes as a messenger and effector molecule. The physiological importance of NO is the result of its high affinity for iron in the active sites of proteins such as guanylate cyclase. Indeed, NO possesses a rich coordination chemistry with iron and the formation of dinitrosyl-dithiolato iron complexes (DNICs) is well documented. In mammals, NO generated by cytotoxic activated macrophages has been reported to play a role as a cytotoxic effector against tumor cells by binding and releasing intracellular iron. Studies from our laboratory have shown that two proteins traditionally involved in drug resistance, namely multidrug-resistance protein 1 and glutathione S-transferase, play critical roles in intracellular NO transport and storage through their interaction with DNICs (R.N. Watts et al., Proc. Natl. Acad. Sci. USA 103:7670-7675, 2006; H. Lok et al., J. Biol. Chem. 287:607-618, 2012). Notably, DNICs are present at high concentrations in cells and are biologically available. These complexes have a markedly longer half-life than free NO, making them an ideal "common currency" for this messenger molecule. Considering the many critical roles NO plays in health and disease, a better understanding of its intracellular trafficking mechanisms will be vital for the development of new therapeutics.
Collapse
Affiliation(s)
- H C Lok
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - S Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - V Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Z Kovacevic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - D R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
13
|
Synthesis, characterization, and fiber-optic infrared reflectance spectroelectrochemical studies of some dinitrosyl iron diphosphine complexes Fe(NO)2L2 (L = P(C6H4X)3). J Organomet Chem 2014. [DOI: 10.1016/j.jorganchem.2013.12.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
Costa E, Faiad O, Landgraf R, Ferreira A, Brigatte P, Curi R, Cury Y, Sampaio S. Involvement of formyl peptide receptors in the stimulatory effect of crotoxin on macrophages co-cultivated with tumour cells. Toxicon 2013; 74:167-78. [DOI: 10.1016/j.toxicon.2013.08.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 06/27/2013] [Accepted: 08/13/2013] [Indexed: 12/01/2022]
|
15
|
Mkonyi LE, Bletsa A, Bolstad AI, Bakken V, Wiig H, Berggreen E. Gingival lymphatic drainage protects against Porphyromonas gingivalis-induced bone loss in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:907-16. [PMID: 22901755 DOI: 10.1016/j.ajpath.2012.05.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 05/08/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
Periodontitis is characterized by tissue destruction and bone loss mainly due to inflammatory responses after bacterial challenge of the gingiva. Gingiva is supplied with lymphatics that drain interstitial fluid and transport immune cells to the lymph nodes for antigen presentation; yet, the role of lymphatics in periodontal disease development is unknown. To investigate the lymphatic function after periodontal infection, we used K14-VEGF receptor 3-Ig (K14) mice that lack lymphatics in gingiva. Mice were orally infected with human Porphyromonas gingivalis and observed for 42 days. The infected K14 mice developed significantly more bone loss than the wild-type mice, and were associated with an increased number of macrophages and major histocompatibility complex class II antigen-presenting cells in the bone resorptional areas. The infected transgenic mice expressed a significant higher periodontal level of several proinflammatory cytokines, whereas the plasma level of P. gingivalis IgG was significantly lower than in the wild-type mice. No differences were found in immune cell distribution in draining lymph nodes between the strains. Our results show that a strong periodontal inflammatory response and a weakened systemic humoral B-cell response took place in K14 mice after infection. We conclude that gingival lymphatics protect against P. gingivalis-induced periodontitis, and we speculate that they are critical in the protection by clearance of infection and by promotion of humoral immune responses.
Collapse
|
16
|
Management of the virulent influenza virus infection by oral formulation of nonhydrolized carnosine and isopeptide of carnosine attenuating proinflammatory cytokine-induced nitric oxide production. Am J Ther 2012; 19:e25-47. [PMID: 20841992 DOI: 10.1097/mjt.0b013e3181dcf589] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Inducible nitric oxide synthase (iNOS) plays an important role in mediating inflammation. In our studies, we found that iNOS-derived NO was significantly increased in the serum samples of 150 patients infected with influenza A virus in comparison with samples of 140 healthy individuals. In human lung epithelial cells, infection with influenza A virus or stimulation with poly(I:C) + interferon-gamma resulted in increased mRNA and protein levels of both interleukin-32 and iNOS, with subsequent release of NO. Activated macrophages are also a source of nitric oxide (NO), which is largely produced by iNOS in response to proinflammatory cytokines. In this review article, the presented findings have many important implications for understanding the Influenza A (H1N1) viral pathogenesis, prevention, and treatment. The direct viral cytotoxicity (referred cytopathic effect) is only a fraction of several types of events induced by virus infection. Nitric oxide and oxygen free radicals such as superoxide anion (O₂⁻˙) are generated markedly in influenza A (including H1N1) virus-infected host boosts, and these molecular species are identified as the potent pathogenic agents. The mutual interaction of NO with O₂⁻˙ resulting in formation of peroxynitrite is operative in the pathogenic mechanism of influenza virus pneumonia. The toxicity and reactivity of oxygen radicals, generated in excessive amounts mediate the overreaction of the host's immune response against the organs or tissues in which viruses are replicating, and this may explain the mechanism of tissue injuries observed in influenza virus infection of various types. The authors revealed the protection that carnosine and its bioavailable nonhydrolized forms provide against peroxynitrite damage and other types of viral injuries in which immunologic interactions are usually involved. Carnosine (beta-alanyl-L-histidine) shows the pharmacologic intracellular correction of NO release which might be one of the important factors of natural immunity in controlling the initial stages of influenza A virus infection (inhibition of virus replication) and virus-induced regulation of cytokine gene expression. The protective effects of orally applied nonhydrolized formulated species of carnosine include at least direct interaction with nitric oxide, inhibition of cytotoxic NO-induced proinflammatory condition, and attenuation of the effects of cytokines and chemokines that can exert profound effects on inflammatory cells. These data are consistent with the hypothesis that natural products, such as chicken soup and chicken breast extracts rich in carnosine and its derivative anserine (beta-alanyl-1-methyl-L-histidine) could contribute to the pathogenesis and prevention of influenza virus infections and cold but have a limitation due to susceptibility to enzymatic hydrolysis of dipeptides with serum carnosinase and urine excretion after oral ingestion of a commercial chicken extract. The developed and patented by the authors formulations of nonhydrolized in digestive tract and blood natural carnosine peptide and isopeptide (gamma-glutamyl-carnosine) products have a promise in the Influenza A (H1N1) virus infection disease control and prevention.
Collapse
|
17
|
Suryo Rahmanto Y, Kalinowski DS, Lane DJR, Lok HC, Richardson V, Richardson DR. Nitrogen monoxide (NO) storage and transport by dinitrosyl-dithiol-iron complexes: long-lived NO that is trafficked by interacting proteins. J Biol Chem 2012; 287:6960-8. [PMID: 22262835 DOI: 10.1074/jbc.r111.329847] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nitrogen monoxide (NO) markedly affects intracellular iron metabolism, and recent studies have shown that molecules traditionally involved in drug resistance, namely GST and MRP1 (multidrug resistance-associated protein 1), are critical molecular players in this process. This is mediated by interaction of these proteins with dinitrosyl-dithiol-iron complexes (Watts, R. N., Hawkins, C., Ponka, P., and Richardson, D. R. (2006) Proc. Natl. Acad. Sci. U.S.A. 103, 7670-7675; Lok, H. C., Suryo Rahmanto, Y., Hawkins, C. L., Kalinowski, D. S., Morrow, C. S., Townsend, A. J., Ponka, P., and Richardson, D. R. (2012) J. Biol. Chem. 287, 607-618). These complexes are bioavailable, have a markedly longer half-life compared with free NO, and form in cells after an interaction between iron, NO, and glutathione. The generation of dinitrosyl-dithiol-iron complexes acts as a common currency for NO transport and storage by MRP1 and GST P1-1, respectively. Understanding the biological trafficking mechanisms involved in the metabolism of NO is vital for elucidating its many roles in cellular signaling and cytotoxicity and for development of new therapeutic targets.
Collapse
Affiliation(s)
- Yohan Suryo Rahmanto
- Department of Pathology, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Henkel A, Kather N, Mönch B, Northoff H, Jauch J, Werz O. Boswellic acids from frankincense inhibit lipopolysaccharide functionality through direct molecular interference. Biochem Pharmacol 2011; 83:115-21. [PMID: 22001311 DOI: 10.1016/j.bcp.2011.09.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 09/24/2011] [Accepted: 09/28/2011] [Indexed: 10/17/2022]
Abstract
Lipophilic extracts of gum resins of Boswellia species (BSE) are used in folk medicine to treat various inflammatory disorders and infections. The molecular background of the beneficial pharmacological effects of such extracts is still unclear. Various boswellic acids (BAs) have been identified as abundant bioactive ingredients of BSE. Here we report the identification of defined BAs as direct inhibitors of lipopolysaccharide (LPS) functionality and LPS-induced cellular responses. In pull-down experiments, LPS could be precipitated using an immobilized BA, implying direct molecular interactions. Binding of BAs to LPS leads to an inhibition of LPS activity which was observed in vitro using a modified limulus amoebocyte lysate assay. Analysis of different BAs revealed clear structure-activity relationships with the classical β-BA as most potent derivative (IC(50)=1.8 μM). In RAW264.7 cells, LPS-induced expression of inducible nitric oxide synthase (iNOS, EC 1.14.13.39) was selectively inhibited by those BAs that interfered with LPS activity. In contrast, interferon-γ-induced iNOS induction was not affected by BAs. We conclude that structurally defined BAs are LPS inhibiting agents and we suggest that β-BA may contribute to the observed anti-inflammatory effects of BSE during infections by suppressing LPS activity.
Collapse
Affiliation(s)
- Arne Henkel
- Department for Pharmaceutical Analytics, Pharmaceutical Institute, University of Tuebingen, Germany.
| | | | | | | | | | | |
Collapse
|
19
|
Vanin AF. Prospects of using magnetic nanoparticles to potentiate the anticarcinogenic action of dinitrosyl iron complexes with thiol ligands. Biophysics (Nagoya-shi) 2011. [DOI: 10.1134/s0006350911050228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Vanin AF, Chazov EI. Prospects of designing medicines with diverse therapeutic activity on the basis of dinitrosyl iron complexes with thiol-containing ligands. Biophysics (Nagoya-shi) 2011. [DOI: 10.1134/s0006350911020321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
21
|
Lewandowska H, Kalinowska M, Brzóska K, Wójciuk K, Wójciuk G, Kruszewski M. Nitrosyl iron complexes--synthesis, structure and biology. Dalton Trans 2011; 40:8273-89. [PMID: 21643591 DOI: 10.1039/c0dt01244k] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nitrosyl complexes of iron are formed in living species in the presence of nitric oxide. They are considered a form in which NO can be stored and stabilized within a living cell. Upon entering a topic in bioinorganic chemistry the researcher faces a wide spectrum of issues concerning synthetic methods, the structure and chemical properties of the complex on the one hand, and its biological implications on the other. The aim of this review is to present the newest knowledge on nitrosyl iron complexes, summarizing the issues that are important for understanding the nature of nitrosyl iron complexes, their possible interactions, behavior in vitro and in vivo, handling of the preparations etc. in response to the growing interest in these compounds. Herein we focus mostly on the dinitrosyl iron complexes (DNICs) due to their prevailing occurrence in NO-treated biological samples. This article reviews recent knowledge on the structure, chemical properties and biological action of DNICs and some mononitrosyls of heme proteins. Synthetic methods are also briefly reviewed.
Collapse
Affiliation(s)
- Hanna Lewandowska
- Institute of Nuclear Chemistry and Technology, Centre for Radiobiology and Biological Dosimetry, 16 Dorodna Str., 03-195, Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
22
|
Vanin AF. Dinitrosyl iron complexes with thiolate ligands: Physico-chemistry, biochemistry and physiology. Nitric Oxide 2009; 21:1-13. [DOI: 10.1016/j.niox.2009.03.005] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 03/23/2009] [Accepted: 03/31/2009] [Indexed: 10/20/2022]
|
23
|
Oseko F, Yamamoto T, Akamatsu Y, Kanamura N, Iwakura Y, Imanishi J, Kita M. IL-17 is involved in bone resorption in mouse periapical lesions. Microbiol Immunol 2009; 53:287-94. [DOI: 10.1111/j.1348-0421.2009.00123.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
24
|
Toledo JC, Bosworth CA, Hennon SW, Mahtani HA, Bergonia HA, Lancaster JR. Nitric oxide-induced conversion of cellular chelatable iron into macromolecule-bound paramagnetic dinitrosyliron complexes. J Biol Chem 2008; 283:28926-33. [PMID: 18480062 DOI: 10.1074/jbc.m707862200] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
One of the most important biological reactions of nitric oxide (nitrogen monoxide, *NO) is its reaction with transition metals, of which iron is the major target. This is confirmed by the ubiquitous formation of EPR-detectable g=2.04 signals in cells, tissues, and animals upon exposure to both exogenous and endogenous *NO. The source of the iron for these dinitrosyliron complexes (DNIC), and its relationship to cellular iron homeostasis, is not clear. Evidence has shown that the chelatable iron pool (CIP) may be at least partially responsible for this iron, but quantitation and kinetic characterization have not been reported. In the murine cell line RAW 264.7, *NO reacts with the CIP similarly to the strong chelator salicylaldehyde isonicotinoyl hydrazone (SIH) in rapidly releasing iron from the iron-calcein complex. SIH pretreatment prevents DNIC formation from *NO, and SIH added during the *NO treatment "freezes" DNIC levels, showing that the complexes are formed from the CIP, and they are stable (resistant to SIH). DNIC formation requires free *NO, because addition of oxyhemoglobin prevents formation from either *NO donor or S-nitrosocysteine, the latter treatment resulting in 100-fold higher intracellular nitrosothiol levels. EPR measurement of the CIP using desferroxamine shows quantitative conversion of CIP into DNIC by *NO. In conclusion, the CIP is rapidly and quantitatively converted to paramagnetic large molecular mass DNIC from exposure to free *NO but not from cellular nitrosothiol. These results have important implications for the antioxidative actions of *NO and its effects on cellular iron homeostasis.
Collapse
Affiliation(s)
- José C Toledo
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
25
|
Richardson DR, Lok HC. The nitric oxide–iron interplay in mammalian cells: Transport and storage of dinitrosyl iron complexes. Biochim Biophys Acta Gen Subj 2008; 1780:638-51. [DOI: 10.1016/j.bbagen.2007.12.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/03/2007] [Accepted: 12/18/2007] [Indexed: 02/05/2023]
|
26
|
Tripathi A, Sodhi A. Production of nitric oxide by murine peritoneal macrophages in vitro on treatment with prolactin and growth hormone: Involvement of protein tyrosine kinases, Ca++, and MAP kinase signal transduction pathways. Mol Immunol 2007; 44:3185-94. [PMID: 17336385 DOI: 10.1016/j.molimm.2007.01.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/22/2007] [Accepted: 01/25/2007] [Indexed: 11/25/2022]
Abstract
Prolactin (PRL) and growth hormone (GH) (somatotropin) have been known to possess immunomodulatory properties. In the present studies we have investigated the production of nitric oxide (NO) and TNF-alpha by murine peritoneal macrophages in vitro on treatment with PRL and GH and the signal transduction mechanism involved. It is observed that significantly enhanced production of NO is induced in macrophages on treatment with PRL and GH. It is further observed that protein tyrosine kinases, MAP kinases and Ca(++) channeling are involved in NO production by macrophages on in vitro treatment with PRL and GH. GH and PRL induced nitric oxide did not have any effect on the expression and production of TNF-alpha. PRL or GH induced TNF-alpha production by murine macrophages was insensitive in the presence of competitive inhibitor of NOS, L-NMMA. Similarly, there is no autocrine or paracrine effect of TNF-alpha on GH or PRL induced NO production and iNOS expression.
Collapse
Affiliation(s)
- Anurag Tripathi
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi-221005, India
| | | |
Collapse
|
27
|
Kleschyov AL, Wenzel P, Munzel T. Electron paramagnetic resonance (EPR) spin trapping of biological nitric oxide. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 851:12-20. [PMID: 17070113 DOI: 10.1016/j.jchromb.2006.10.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Revised: 10/05/2006] [Accepted: 10/06/2006] [Indexed: 10/24/2022]
Abstract
Nitric oxide (NO) is a free radical species with multiple physiological functions. Because of low concentrations and short half-life of NO, its direct measurement in living tissues remains a difficult task. Electron paramagnetic resonance (EPR) spin trapping is probably one of the best suitable platforms for development of new methods for quantification of biological NO. The most reliable EPR-based approaches developed so far are based on the reaction of NO with various iron complexes, both intrinsic and exogenously applied. This review is focused on the current state and perspectives of EPR spin trapping for experimental and clinical NO biology.
Collapse
Affiliation(s)
- Andrei L Kleschyov
- Second Department of Medicine, Johannes Gutenberg University of Mainz, Mainz 55131, Germany.
| | | | | |
Collapse
|
28
|
Pedersen JZ, De Maria F, Turella P, Federici G, Mattei M, Fabrini R, Dawood KF, Massimi M, Caccuri AM, Ricci G. Glutathione Transferases Sequester Toxic Dinitrosyl-Iron Complexes in Cells. J Biol Chem 2007; 282:6364-71. [PMID: 17197702 DOI: 10.1074/jbc.m609905200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It is now well established that exposure of cells and tissues to nitric oxide leads to the formation of a dinitrosyl-iron complex bound to intracellular proteins, but little is known about how the complex is formed, the identity of the proteins, and the physiological role of this process. By using EPR spectroscopy and enzyme activity measurements to study the mechanism in hepatocytes, we here identify the complex as a dinitrosyl-diglutathionyl-iron complex (DNDGIC) bound to Alpha class glutathione S-transferases (GSTs) with extraordinary high affinity (K(D) = 10(-10) m). This complex is formed spontaneously through NO-mediated extraction of iron from ferritin and transferrin, in a reaction that requires only glutathione. In hepatocytes, DNDGIC may reach concentrations of 0.19 mm, apparently entirely bound to Alpha class GSTs, present in the cytosol at a concentration of about 0.3 mm. Surprisingly, about 20% of the dinitrosyl-glutathionyl-iron complex-GST is found to be associated with subcellular components, mainly the nucleus, as demonstrated in the accompanying paper (Stella, L., Pallottini, V., Moreno, S., Leoni, S., De Maria, F., Turella, P., Federici, G., Fabrini, R., Dawood, K. F., Lo Bello, M., Pedersen, J. Z., and Ricci, G. (2007) J. Biol. Chem. 282, 6372-6379). DNDGIC is a potent irreversible inhibitor of glutathione reductase, but the strong complex-GST interaction ensures full protection of glutathione reductase activity in the cells, and in vitro experiments show that damage to the reductase only occurs when the DNDGIC concentration exceeds the binding capacity of the intracellular GST pool. Because Pi class GSTs may exert a similar role in other cell types, we suggest that specific sequestering of DNDGIC by GSTs is a physiological protective mechanism operating in conditions of excessive levels of nitric oxide.
Collapse
Affiliation(s)
- Jens Z Pedersen
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Children's Hospital Bambin Gesù, 00165 Rome
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gonzalez A, Restrepo A, Cano LE. Role of iron in the nitric oxide-mediated fungicidal mechanism of IFN-gamma-activated murine macrophages against Paracoccidioides brasiliensis conidia. Rev Inst Med Trop Sao Paulo 2007; 49:11-6. [PMID: 17384814 DOI: 10.1590/s0036-46652007000100003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Accepted: 08/01/2006] [Indexed: 11/21/2022] Open
Abstract
Iron is an essential growth element of virtually all microorganisms and its restriction is one of the mechanisms used by macrophages to control microbial multiplication. Paracoccidioides brasiliensis, the agent of paracoccidioidomycosis, an important systemic mycosis in Latin America, is inhibited in its conidia-to-yeast conversion in the absence of iron. We studied the participation of iron in the nitric oxide (NO)-mediated fungicidal mechanism against conidia. Peritoneal murine macrophages activated with 50U/mL of IFN-gamma or treated with 35 µM Deferoxamine (DEX) and infected with P. brasiliensis conidia, were co-cultured and incubated for 96 h in the presence of different concentrations of holotransferrin (HOLO) and FeS0(4). The supernatants were withdrawn in order to assess NO2 production by the Griess method. The monolayers were fixed, stained and observed microscopically. The percentage of the conidia-to-yeast transition was estimated by counting 200 intracellular propagules. IFN-gamma-activated or DEX-treated Mthetas presented marked inhibition of the conidia-to-yeast conversion (19 and 56%, respectively) in comparison with non-activated or untreated Mthetas (80%). IFN-gamma-activated macrophages produced high NO levels in comparison with the controls. Additionally, when the activated or treated-macrophages were supplemented with iron donors (HOLO or FeSO4), the inhibitory action was reversed, although NO production remained intact. These results suggest that the NO-mediated fungicidal mechanism exerted by IFN-gamma-activated macrophages against P. brasiliensis conidia, is dependent of an iron interaction.
Collapse
Affiliation(s)
- Angel Gonzalez
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas, Medellín, Colombia.
| | | | | |
Collapse
|
30
|
Kesherwani V, Sodhi A. Differential activation of macrophages in vitro by lectin Concanavalin A, Phytohemagglutinin and Wheat germ agglutinin: production and regulation of nitric oxide. Nitric Oxide 2006; 16:294-305. [PMID: 17208474 DOI: 10.1016/j.niox.2006.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 11/15/2006] [Accepted: 11/16/2006] [Indexed: 10/23/2022]
Abstract
The role of Concanavalin A (ConA), Phytohemagglutinin (PHA) and Wheat germ agglutinin (WGA) in the activation of murine peritoneal macrophages particularly with reference to production and regulation of nitric oxide (NO) has been investigated. Macrophages on treatment with ConA and PHA showed significantly enhanced production of NO, which was dose and time dependent. On the other hand macrophages treated with WGA did not produce NO. L-N-monomethyal-l-arginine (L-NMMA), an inhibitor of NOS inhibited the ConA and PHA induced NO production. ConA and PHA treatment of macrophages induced transcription of iNOS gene and the enhanced expression of iNOS protein. Pharmacological inhibitors of PI3 kinase-Wortmannin, tyrosine kinase-Genestein, protein kinase C-H-7 and p42/44-PD98059 inhibited the ConA and PHA induced production of NO and p38 MAP kinase inhibitor SB202190 inhibited NO production only in ConA treated macrophage, while Galphai protein inhibitor-PTX and JNK inhibitor-SP600125 inhibited NO production in PHA treated macrophages. Tyrophostin (AG490), an inhibitor of JAK2 and TMB-8, an intracellular calcium immobilizing agent also inhibited the ConA and PHA induced NO production, suggesting the involvement of JAK-STAT pathway and calcium. The data also provides the relative measure and importance of different key signaling molecules in the regulation of NO production by macrophages on activation.
Collapse
Affiliation(s)
- Varun Kesherwani
- School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi 221005, India
| | | |
Collapse
|
31
|
Stuehr DJ, Griffith OW. Mammalian nitric oxide synthases. ADVANCES IN ENZYMOLOGY AND RELATED AREAS OF MOLECULAR BIOLOGY 2006; 65:287-346. [PMID: 1373932 DOI: 10.1002/9780470123119.ch8] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- D J Stuehr
- Department of Medicine, Cornell University Medical College, New York, New York
| | | |
Collapse
|
32
|
Knott EM, Sun J, Lei Y, Ryou MG, Olivencia-Yurvati AH, Mallet RT. Pyruvate mitigates oxidative stress during reperfusion of cardioplegia-arrested myocardium. Ann Thorac Surg 2006; 81:928-34. [PMID: 16488697 DOI: 10.1016/j.athoracsur.2005.08.046] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Revised: 08/24/2005] [Accepted: 08/25/2005] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cardioplegic arrest and reperfusion of the myocardium imposes oxidative stress that could potentially inactivate metabolic enzymes and compromise energy production. This study determined the impact of cardioplegic arrest and reperfusion on activities of several oxidant-sensitive enzymes, and tested whether pyruvate, a natural metabolic fuel and antioxidant, mitigates oxidant stress, protects enzymes, and bolsters myocardial energy state after reperfusion. METHODS In situ swine hearts were arrested for 60 minutes with 4:1 blood:crystalloid cardioplegia, and then reperfused for 3 minutes with cardioplegia-free blood with or without approximately 12 mM pyruvate. Tissue metabolites and enzyme activities were measured in left ventricular myocardium snap frozen at 45 minutes of arrest and 3 minutes of reperfusion. RESULTS The 8-isoprostane content, a measure of lipid peroxidation, sharply increased upon reperfusion, coincident with a 70% decline in redox state of the intracellular antioxidant glutathione. Aconitase and glucose 6-phosphate dehydrogenase activities fell during arrest; creatine kinase and phosphofructokinase were inactivated upon reperfusion. Pyruvate suppressed 8-isoprostane formation, maintained glutathione redox state, and enhanced phosphocreatine phosphorylation potential, a measure of myocardial energy state, during reperfusion. Pyruvate reactivated creatine kinase and aconitase, which are at least partially mitochondrial enzymes, but did not protect the cytosolic enzymes glucose 6-phosphate dehydrogenase and phosphofructokinase. CONCLUSIONS Administration of pyruvate upon reperfusion after cardioplegic arrest mitigates oxidative stress, protects mitochondrial enzymes and increases myocardial energy state. These results support therapeutic application of pyruvate-enhanced reperfusion to prevent cardiac injury after cardioplegic arrest.
Collapse
Affiliation(s)
- E Marty Knott
- Department of Integrative Physiology, University of North Texas Health Science Center, Fort Worth, Texas 76107-2699, USA
| | | | | | | | | | | |
Collapse
|
33
|
Watts RN, Hawkins C, Ponka P, Richardson DR. Nitrogen monoxide (NO)-mediated iron release from cells is linked to NO-induced glutathione efflux via multidrug resistance-associated protein 1. Proc Natl Acad Sci U S A 2006; 103:7670-5. [PMID: 16679408 PMCID: PMC1472503 DOI: 10.1073/pnas.0602515103] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nitrogen monoxide (NO) plays a role in the cytotoxic mechanisms of activated macrophages against tumor cells by inducing iron (Fe) release. We have shown that NO-mediated Fe efflux from cells required glutathione (GSH), and we have hypothesized that a GS-Fe-NO complex was released. Hence, we studied the role of the GSH-conjugate transporter multidrug resistance-associated protein 1 (MRP1) in NO-mediated Fe efflux. MCF7-VP cells overexpressing MRP1 exhibited a 3- to 4-fold increase in NO-mediated 59Fe and GSH efflux compared with WT cells (MCF7-WT) over 4 h. Similar results were found for other MRP1-overexpressing cell types but not those expressing another drug efflux pump, P-glycoprotein. NO-mediated 59Fe and GSH efflux were temperature- and energy-dependent and were significantly decreased by the GSH-depleting agent and MRP1 transport inhibitor L-buthionine-[S,R]-sulfoximine. Other MRP1 inhibitors, MK571, probenecid, and difloxacin, significantly inhibited NO-mediated 59Fe release. EPR spectroscopy demonstrated the dinitrosyl-dithiol-Fe complex (DNIC) peak in NO-treated cells was increased by MRP1 inhibitors, indicating inhibited DNIC transport from cells. The extent of DNIC accumulation correlated with the ability of MRP1 inhibitors to prevent NO-mediated 59Fe efflux. MCF7-VP cells were more sensitive than MCF7-WT cells to growth inhibition by effects of NO, which was potentiated by L-buthionine-[S,R]-sulfoximine. These data indicate the importance of GSH in NO-mediated inhibition of proliferation. Collectively, NO stimulates Fe and GSH efflux from cells via MRP1. Active transport of NO by MRP1 overcomes diffusion that is inefficient and nontargeted, which has broad ramifications for understanding NO biology.
Collapse
Affiliation(s)
- Ralph N. Watts
- *Children's Cancer Institute Australia for Medical Research, Sydney 2031, Australia
| | - Clare Hawkins
- The Heart Research Institute, Sydney 2050, Australia; and
| | - Prem Ponka
- Lady Davis Institute, Jewish General Hospital and Department of Physiology, McGill University, Montreal, QC, Canada H3T 1E2
- To whom correspondence may be addressed. E-mail:
or
| | - Des R. Richardson
- *Children's Cancer Institute Australia for Medical Research, Sydney 2031, Australia
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
34
|
Abstract
The immune system is involved at all stages of the atherosclerotic disease process. Innate immunity, represented by macrophages and other cells, is directly activated by microbial components and possibly also by autologous lipids and proteins. It elicits inflammatory activity, which is a key component of the atherosclerotic lesion. Adaptive immunity is initiated by recognition of disease-related antigens, which include oxidatively modified lipoproteins, heat shock proteins and microbial macromolecules. In the artery wall, adaptive immune recognition mainly leads to Thl effector responses, which are characterized by secretion of proinflammatory cytokines and by activation of macrophages and vascular cells. Therefore, both the innate and adaptive arms of the immune system lead to inflammation in the developing atherosclerotic lesion. Interestingly, several effector pathways of cellular as well as humoral immunity tend to counteract proatherogenic, proinflammatory immunity. The notion that immunity plays an important role in the development of atherosclerosis has focused attention on a number of potential novel targets for intervention based on modulation of such immune responses.
Collapse
Affiliation(s)
- G K Hansson
- Centre for Molecular Medicine and Department of Medicine, Karolinska Institute, Stockholm, Sweden.
| | | |
Collapse
|
35
|
Sodhi A, Sharma RK, Batra HV, Tuteja U. Recombinant fraction 1 protein of Yersinia pestis activates murine peritoneal macrophages in vitro. Cell Immunol 2004; 229:52-61. [PMID: 15331328 DOI: 10.1016/j.cellimm.2004.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Accepted: 05/19/2004] [Indexed: 01/04/2023]
Abstract
Fraction 1 antigen of Yersinia pestis is a capsule protein of 17.5kDa, known to induce thymocyte proliferation and have anti-phagocytic role in macrophages. It serves as a major protective antigen against challenge of Y. pestis by inducing high concentration of IgG1 antibody response. In the present investigation it is observed that 10microg/ml of rF1 antigen activated murine peritoneal macrophages in vitro. rF1 induced the production of TNF-alpha, IL-1, IL-6, and NO. rF1 treatment also induced increased transcription of IFN-gamma and its related chemokines KC, IP-10, MIP-1alpha, MIP-1beta, MCP-1, RANTES in macrophages. Significantly increased transcription of TLR5 was observed in macrophages treated with rF1, while the expression of TLR2 and TLR4 remained unaffected.
Collapse
Affiliation(s)
- Ajit Sodhi
- School of Biotechnology, Banaras Hindu University, Varanasi 221 005, UP, India.
| | | | | | | |
Collapse
|
36
|
Sasaki H, Balto K, Kawashima N, Eastcott J, Hoshino K, Akira S, Stashenko P. Gamma interferon (IFN-gamma) and IFN-gamma-inducing cytokines interleukin-12 (IL-12) and IL-18 do not augment infection-stimulated bone resorption in vivo. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2004; 11:106-10. [PMID: 14715554 PMCID: PMC321357 DOI: 10.1128/cdli.11.1.106-110.2004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Periapical granulomas are induced by bacterial infection of the dental pulp and result in destruction of the surrounding alveolar bone. In previous studies we have reported that the bone resorption in this model is primarily mediated by macrophage-expressed interleukin-1 (IL-1). The expression and activity of IL-1 is in turn modulated by a network of Th1 and Th2 regulatory cytokines. In the present study, the functional roles of the Th1 cytokine gamma interferon (IFN-gamma) and IFN-gamma-inducing cytokines IL-12 and IL-18 were determined in a murine model of periapical bone destruction. IL-12-/-, IL-18-/-, and IFN-gamma-/- mice were subjected to surgical pulp exposure and infection with a mixture of four endodontic pathogens, and bone destruction was determined by microcomputed tomography on day 21. The results indicated that all IL-12-/-, IL-18-/-, and IFN-gamma-/- mice had similar infection-stimulated bone resorption in vivo as wild-type control mice. Mice infused with recombinant IL-12 also had resorption similar to controls. IFN-gamma-/- mice exhibited significant elevations in IL-6, IL-10, IL-12, and tumor necrosis factor alpha in lesions compared to wild-type mice, but these modulations had no net effect on IL-1alpha levels. Recombinant IL-12, IL-18, and IFN-gamma individually failed to consistently modulate macrophage IL-1alpha production in vitro. We conclude that, at least individually, endogenous IL-12, IL-18, and IFN-gamma do not have a significant effect on the pathogenesis of infection-stimulated bone resorption in vivo, suggesting possible functional redundancy in proinflammatory pathways.
Collapse
Affiliation(s)
- Hajime Sasaki
- Department of Cytokine Biology, The Forsyth Institute, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Sinbandhit-Tricot S, Cillard J, Chevanne M, Morel I, Cillard P, Sergent O. Glutathione depletion increases nitric oxide-induced oxidative stress in primary rat hepatocyte cultures: involvement of low-molecular-weight iron. Free Radic Biol Med 2003; 34:1283-94. [PMID: 12726916 DOI: 10.1016/s0891-5849(03)00108-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Various drugs and chemicals can cause a glutathione (GSH) depletion in the liver. Moreover, nitric oxide (NO) can be generated in response to physiological and pathological situations such as inflammation. The aim of this study was to estimate oxidative stress when primary rat hepatocytes were exposed to GSH depletion after NO production. For this purpose, cells were preincubated with lipopolysaccharide (LPS) and gamma-interferon (IFN) for 18 h in order to induce NO production by NO synthase and then L-buthionine sulfoximine (BSO), an inhibitor of GSH synthesis, was added for 5 h. In hepatocyte cultures preincubated with LPS and IFN before BSO addition, an increase in lipid peroxidation was noted. In those cells, an elevation of iron-bound NO and a decrease in free NO led us to suggest the involvement of low-molecular-weight iron (LMW iron) in the enhancement of oxidative stress. Indeed, addition of deferiprone, a chelator of LMW iron, reduced iron-bound NO levels and the extent of oxidative stress. Moreover, an important elevation of LMW iron levels was also observed. As both, N-acetylcysteine, a GSH precursor, and N(G)-monomethyl-L-arginine, a NO synthase inhibitor, totally inhibited the elevation of LMW iron and oxidative stress, a cooperative role could be attributed to NO production and GSH depletion.
Collapse
|
38
|
Pieper GM, Halligan NLN, Hilton G, Konorev EA, Felix CC, Roza AM, Adams MB, Griffith OW. Non-heme iron protein: a potential target of nitric oxide in acute cardiac allograft rejection. Proc Natl Acad Sci U S A 2003; 100:3125-30. [PMID: 12624190 PMCID: PMC152257 DOI: 10.1073/pnas.0636938100] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We examined iron nitrosylation of non-heme protein and enzymatic activity of the Fe-S cluster protein, aconitase, in acute cardiac allograft rejection. Heterotopic transplantation of donor hearts was performed in histocompatibility matched (isografts: Lewis --> Lewis) and mismatched (allografts: Wistar-Furth --> Lewis) rats. On postoperative days (POD) 4-6, Western blot analysis and immunohistochemistry revealed inducible nitric-oxide synthase (iNOS) protein in allografts but not isografts. EPR spectroscopy revealed background signals at g = 2.003 (for semiquinone) and g = 2.02 and g = 1.94 (for Fe-S cluster protein) in isografts and normal hearts. In contrast, in allografts on POD4, a new axial signal at g = 2.04 and g = 2.02 appeared that was attributed to the dinitrosyl-iron complex formed by nitrosylation of non-heme protein. Appearance of this signal occurred at or before significant nitrosylation of heme protein. Iron nitrosylation of non-heme protein was coincidental with decreases in the nonnitrosylated Fe-S cluster signal at g = 1.94. Aconitase enzyme activity was decreased to approximately 50% of that observed in isograft controls by POD4. Treatment with cyclosporine blocked the (i) elevation of plasma nitrate + nitrite, (ii) up-regulation of iNOS protein, (iii) decrease in Fe-S cluster EPR signal, (iv) formation of dinitrosyl-iron complexes, and (v) loss of aconitase enzyme activity. Formation of dinitrosyl-iron complexes and loss of aconitase activity within allografts also was inhibited by treatment of recipients with a selective iNOS inhibitor, l-N(6)-(1-iminoethyl)lysine. This report shows targeting of an important non-heme Fe-S cluster protein in acute solid organ transplant rejection.
Collapse
Affiliation(s)
- Galen M Pieper
- Department of Surgery, Division of Transplant Surgery, Medical College of Wisconsin, 9200 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Watts RN, Richardson DR. The mechanism of nitrogen monoxide (NO)-mediated iron mobilization from cells. NO intercepts iron before incorporation into ferritin and indirectly mobilizes iron from ferritin in a glutathione-dependent manner. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:3383-92. [PMID: 12135476 DOI: 10.1046/j.1432-1033.2002.02987.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Nitrogen monoxide (NO) is a cytotoxic effector molecule produced by macrophages that results in Fe mobilization from tumour target cells which inhibits DNA synthesis and mitochondrial respiration. It is well known that NO has a high affinity for Fe, and we showed that NO-mediated Fe mobilization is markedly potentiated by glutathione (GSH) generated by the hexose monophosphate shunt [Watts, R.N. & Richardson, D.R. (2001) J. Biol. Chem. 276, 4724-4732]. We hypothesized that GSH completes the coordination shell of an NO[bond]Fe complex that is released from the cell. In this report we have extended our studies to further characterize the mechanism of NO-mediated Fe mobilization. Native PAGE 59Fe-autoradiography shows that NO decreased ferritin-59Fe levels in cells prelabelled with [59Fe]transferrin. In prelabelled cells, ferritin-59Fe levels increased 3.5-fold when cells were reincubated with control media between 30 and 240 min. In contrast, when cells were reincubated with NO, ferritin-59Fe levels decreased 10-fold compared with control cells after a 240-min reincubation. However, NO could not remove Fe from ferritin in cell lysates. Our data suggest that NO intercepts 59Fe on route to ferritin, and indirectly facilitates removal of 59Fe from the protein. Studies using the GSH-depleting agent, L-buthionine-(S,R)-sulphoximine, indicated that the reduction in ferritin-59Fe levels via NO was GSH-dependent. Competition experiments with NO and permeable chelators demonstrated that both bind a similar Fe pool. We suggest that NO requires cellular metabolism in order to effect Fe mobilization and this does not occur via passive diffusion down a concentration gradient. Based on our results, we propose a model of glucose-dependent NO-mediated Fe mobilization.
Collapse
Affiliation(s)
- Ralph N Watts
- The Iron Metabolism and Chelation Group, The Heart Research Institute, Camperdown, Sydney, New South Wales, Australia
| | | |
Collapse
|
40
|
Abstract
Sustained TNFalpha induction is central to the pathogenesis of chronic liver disease including alcoholic liver disease (ALD). However, molecular understanding of this abnormality at the cellular level remains elusive. Redox regulation of NF-kappaB is critical in the transcriptional control of TNFalpha expression. Evidence supports that increased iron storage in hepatic macrophages (HM) is causally associated with accentuated and sustained NF-kappaB activation in these cells in ALD. Treatment of cultured HM with a lipophilic iron chelator (deferiprone) abrogates LPS-induced NF-kappaB activation. HM from an animal model of ALD have increased nonheme iron content accompanied by increased generation of EPR-detected radicals, NF-kappaB activation, and TNFalpha induction, all of which are normalized by ex vivo treatment of the cells with deferiprone. A moderate increase in the nonheme iron content in HM by erythrophagocytosis, promotes subsequent LPS-stimulated NF-kappaB activation in a hemeoxygenase-dependent manner. Recent evidence also suggests a role of intracellular low molecular weight iron in the early signal transduction for LPS-mediated NF-kappaB activation.
Collapse
Affiliation(s)
- Hidekazu Tsukamoto
- USC-UCLA Research Center for Alcoholic Liver and Pancreatic Diseases and USC Research Center for Liver Diseases, Keck School of Medicine of University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
41
|
Abstract
The author describes how his experience as an infectious disease fellow at Stanford University with Jack Remington from 1969 to 1971 influenced the direction of his subsequent laboratory investigation. The author reviews a series of studies that were intended to provide a mechanistic understanding of an in vitro cytotoxicity assay developed while he was a fellow with Jack Remington. These investigations resulted in the 1987 discovery of the synthesis of nitric oxide from L-arginine by cytokine-activated macrophages. This work provided the components (the precursor, products, and an inhibitor) of the enzymatic synthesis of nitric oxide by all three later-identified nitric oxide synthase isoforms. The significance of cytokine-induced nitric oxide synthesis during innate resistance and cell-mediated immune reactions is discussed briefly.
Collapse
Affiliation(s)
- John B Hibbs
- Division of Infectious Diseases, Department of Medicine, University of Utah School of Medicine, and VA Medical Center, Salt Lake City, Utah 84132, USA.
| |
Collapse
|
42
|
Ueno T, Suzuki Y, Fujii S, Vanin AF, Yoshimura T. In vivo nitric oxide transfer of a physiological NO carrier, dinitrosyl dithiolato iron complex, to target complex. Biochem Pharmacol 2002; 63:485-93. [PMID: 11853699 DOI: 10.1016/s0006-2952(01)00869-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dinitrosyl dithiolato iron complex (DNIC) has been identified as an endogenous NO carrier, yet in vivo mechanisms of NO donation remain undefined. Transnitrosylation, in which a coordinated NO group is transferred to another metal complex, has been observed in transition-metal-nitrosyl chemistry. In this study, we used three kinds of iron dithiocarbamate complexes (Fe-DTCs) as NO acceptors to elucidate in vivo transnitrosylation of diglutathionyl dinitrosyl iron complex [DNIC-(GS)(2)]. Fe-DTCs were administered to mice after the injection of DNIC-(GS)(2) and electron paramagnetic resonance (EPR) spectra were measured both in the resected organs and in the upper abdomen of living mice. The spectral feature gradually changed from an initial DNIC-(GS)(2) signal to mononitrosyl iron dithiocarbamate one, suggesting that NO-Fe-DTC was formed through in vivo reaction of DNIC-(GS)(2) with Fe-DTC. The spectral results in in vitro and in vivo systems indicate that NO-Fe-DTCs can be formed not only by the transfer of coordinated NO-group(s) in DNIC-(GS)(2) but also by the abstraction of Fe-NO group in DNIC-(GS)(2) by free DTC ligands. Transnitrosylation proceeded more rapidly in blood than in liver and kidney; and more efficiently in kidney than in liver. Further, the ability to accept NO from DNIC was dependent on water-solubility of Fe-DTCs. Thus, in vivo transnitrosylation from DNIC to exogenous iron complex could be observed and this reaction was influenced by biological constituents and properties of iron complex. These results demonstrate that the transnitrosylation from DNIC to intrinsic NO acceptors like metalloproteins has a probable significance in in vivo NO transfer process.
Collapse
Affiliation(s)
- Takaharu Ueno
- Laboratory of Applied Biomedicinal Chemistry, Institute for Life Support Technology, Yamagata Public Corporation for the Development of Industry, Yamagata, Japan
| | | | | | | | | |
Collapse
|
43
|
Biswas SK, Sodhi A, Paul S. Regulation of nitric oxide production by murine peritoneal macrophages treated in vitro with chemokine monocyte chemoattractant protein 1. Nitric Oxide 2001; 5:566-79. [PMID: 11730364 DOI: 10.1006/niox.2001.0370] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Monocyte chemoattractant protein 1 (MCP-1) is an important mediator of monocyte/macrophage recruitment and activation at the sites of chronic inflammation and neoplasia. In the current study, the role of nitrogen monoxide (NO) in the activation of murine peritoneal macrophages to the tumoricidal state in response to in vitro MCP-1 treatment and the regulatory mechanisms involved therein were investigated. Murine peritoneal macrophages upon activation with MCP-1 showed a dose- and time-dependent production of NO together with increased tumoricidal activity against P815 mastocytoma cells. N-monomethyl-l-arginine (L-NMMA), a specific inhibitor of the l-arginine pathway, inhibited the MCP-1-induced NO secretion and generation of macrophage-mediated tumoricidal activity against P815 (NO-sensitive, TNF-resistant) cells but not the L929 (TNF-sensitive, NO-resistant) cells. These results indicated l-arginine-dependent production of NO to be one of the effector mechanisms contributing to the tumoricidal activity of MCP-1-treated macrophages. Supporting this fact, expression of iNOS mRNA was also detected in the murine peritoneal macrophages upon treatment with MCP-1. Investigating the signal transduction pathway responsible for the NO production by the MCP-1-activated murine peritoneal macrophages, it was observed that the pharmacological inhibitors wortmannin, H-7 (1-(5-isoquinoline sulfonyl)-2-methyl piperazine dihydrochloride), and PD98059 blocked the MCP-1-induced NO production, suggesting the probable involvement of phosphoinositol-3-kinase, protein kinase C, and p42/44 MAPkinases in the above process. Various modulators of calcium and calmodulin (CaM) such as EGTA, nifedipine, TMB-8 (3,4,5-trimethoxybenzoic acid-8-(diethylamino)octyl ester), A23187, and W-7 (N-(6-aminohexyl)-5-chloro-1-napthalenesulfonamide) were also found to modulate the in vitro macrophage NO release in response to MCP-1. This observation indicated the regulatory role of calcium/CaM in the process of MCP-1-induced macrophage NO production. Similarly, the role of serine/threonine and protein tyrosine phosphatases in the above pathway was suggested using the specific inhibitors of these phosphatases, okadaic acid and sodium orthovanadate.
Collapse
Affiliation(s)
- S K Biswas
- School of Biotechnology, Banaras Hindu University, Varanasi, 221005, India
| | | | | |
Collapse
|
44
|
Lo Bello M, Nuccetelli M, Caccuri AM, Stella L, Parker MW, Rossjohn J, McKinstry WJ, Mozzi AF, Federici G, Polizio F, Pedersen JZ, Ricci G. Human glutathione transferase P1-1 and nitric oxide carriers; a new role for an old enzyme. J Biol Chem 2001; 276:42138-45. [PMID: 11533048 DOI: 10.1074/jbc.m102344200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
S-Nitrosoglutathione and the dinitrosyl-diglutathionyl iron complex are involved in the storage and transport of NO in biological systems. Their interactions with the human glutathione transferase P1-1 may reveal an additional physiological role for this enzyme. In the absence of GSH, S-nitrosoglutathione causes rapid and stable S-nitrosylation of both the Cys(47) and Cys(101) residues. Ion spray ionization-mass spectrometry ruled out the possibility of S-glutathionylation and confirms the occurrence of a poly-S-nitrosylation in GST P1-1. S-Nitrosylation of Cys(47) lowers the affinity 10-fold for GSH, but this negative effect is minimized by a half-site reactivity mechanism that protects one Cys(47)/dimer from nitrosylation. Thus, glutathione transferase P1-1, retaining most of its original activity, may act as a NO carrier protein when GSH depletion occurs in the cell. The dinitrosyl-diglutathionyl iron complex, which is formed by S-nitrosoglutathione decomposition in the presence of physiological concentrations of GSH and traces of ferrous ions, binds with extraordinary affinity to one active site of this dimeric enzyme (K(i) < 10(-12) m) and triggers negative cooperativity in the vacant subunit (K(i) = 10(-9) m). The complex bound to the enzyme is stable for hours, whereas in the free form and at low concentrations, its life time is only a few minutes. ESR and molecular modeling studies provide a reasonable explanation of this strong interaction, suggesting that Tyr(7) and enzyme-bound GSH could be involved in the coordination of the iron atom. All of the observed findings suggest that glutathione transferase P1-1, by means of an intersubunit communication, may act as a NO carrier under different cellular conditions while maintaining its well known detoxificating activity toward dangerous compounds.
Collapse
Affiliation(s)
- M Lo Bello
- Department of Biology, University of Rome "Tor Vergata," 00133 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Nitric oxide (NO), a simple free radical gas, elicits a surprisingly wide range of physiological and pathophysiological effects. NO interacts with soluble guanylate cyclase to evoke many of these effects. However, NO can also interact with molecular oxygen and superoxide radicals to produce reactive nitrogen species that can modify a number of macromolecules including proteins, lipids, and nucleic acids. NO can also interact directly with transition metals. Here, we have reviewed the non--3',5'-cyclic-guanosine-monophosphate-mediated effects of NO including modifications of proteins, lipids, and nucleic acids.
Collapse
Affiliation(s)
- K L Davis
- Department of Integrated Biology and Pharmacology, University of Texas Houston Health Science Center, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|
46
|
Wang CH, Kuo HP. Nitric oxide modulates interleukin-1beta and tumour necrosis factor-alpha synthesis, and disease regression by alveolar macrophages in pulmonary tuberculosis. Respirology 2001; 6:79-84. [PMID: 11264769 DOI: 10.1046/j.1440-1843.2001.00302.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pretreatment with nitric oxide synthase (NOS) inhibitors profoundly increases mortality, bacterial burden and pathological tissue damage in mice infected with Mycobacterium tuberculosis. Nitric oxide (NO) production is enhanced in alveolar macrophages (AM) of tuberculosis (TB) patients. Interleukin (IL)-1beta and tumour necrosis factor (TNF)-alpha released from AM are involved in the immune response to mycobacterial infection. The aim of the present study was to examine whether NO is implicated in IL-1beta and TNF-alpha synthesis by AM and related to the resolution of disease activity in TB patients. Purified AM were retrieved by bronchoalveolar lavage from TB patients and normal subjects, and cultured in the presence or absence of a NO inhibitor, NG-monomethyl-L-arginine (L-NMMA). The release of IL-1beta and TNF-alpha, and their mRNA expression were determined by enzyme-linked immunosorbent assay (ELISA) and northern analysis, respectively. The level of nitrite released into the culture medium was determined. The rate of disease regression was evaluated by serial chest radiography. The release of nitrite, IL-1beta and TNF-alpha was much greater from AM of TB patients than normal subjects. NG-monomethyl-L-arginine inhibited the production of nitrite as well as IL-1beta and TNF-alpha in TB patients. The mRNA expression for IL-1beta and TNF-alpha was upregulated in TB patients and was depressed by L-NMMA. Immunocytochemistry using a monoclonal antibody against nuclear factor-kappaB (NF-kappaB) subunit p65 showed NF-kappaB was highly expressed and translocated to the nuclei of AM in TB patients, and was inhibited by L-NMMA. An inhibition of NF-kappaB by pyrrolidine dithiocarbamate attenuated IL-1beta and TNF-alpha synthesis. More generation of NO from cultured AM increased the disease regression in TB patients. We conclude that the enhanced NO generation by AM of TB patients may play an autoregulatory role in amplifying the synthesis of pro-inflammatory cytokines, probably through the activation of NF-kappaB. Nitric oxide may also play an important role in resistance to M. tuberculosis infection.
Collapse
Affiliation(s)
- C H Wang
- Department of Thoracic Medicine II, Chang Gung Memorial Hospital, Taipei, Taiwan.
| | | |
Collapse
|
47
|
Wang C, Kuo H. Nitric oxide modulates interleukin‐1β and tumour necrosis factor‐α synthesis, and disease regression by alveolar macrophages in pulmonary tuberculosis. Respirology 2001. [DOI: 10.1111/j.1440-1843.2001.00302.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Chun‐Hua Wang
- Department of Thoracic Medicine II, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Han‐Pin Kuo
- Department of Thoracic Medicine II, Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Watts RN, Richardson DR. Nitrogen monoxide (no) and glucose: unexpected links between energy metabolism and no-mediated iron mobilization from cells. J Biol Chem 2001; 276:4724-32. [PMID: 11078730 DOI: 10.1074/jbc.m006318200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nitrogen monoxide (NO) affects cellular iron metabolism due to its high affinity for this metal ion. Indeed, NO has been shown to increase the mRNA binding activity of the iron-regulatory protein 1, which is a major regulator of iron homeostasis. Recently, we have shown that NO generators increase (59)Fe efflux from cells prelabeled with (59)Fe-transferrin (Wardrop, S. L., Watts, R. N., and Richardson, D. R. (2000) Biochemistry 39, 2748-2758). The mechanism involved in this process remains unknown, and in this investigation we demonstrate that it is potentiated upon adding d-glucose (d-Glc) to the reincubation medium. In d-Glc-free or d-Glc-containing media, 5.6 and 16.5% of cellular (59)Fe was released, respectively, in the presence of S-nitrosoglutathione. This difference in (59)Fe release was observed with a variety of NO generators and cell types and was not due to a change in cell viability. Kinetic studies showed that d-Glc had no effect on the rate of NO production by NO generators. Moreover, only the metabolizable monosaccharides d-Glc and d-mannose could stimulate NO-mediated (59)Fe mobilization, whereas other sugars not easily metabolized by fibroblasts had no effect. Hence, metabolism of the monosaccharides was essential to increase NO-mediated (59)Fe release. Incubation of cells with the citric acid cycle intermediates, citrate and pyruvate, did not enhance NO-mediated (59)Fe release. Significantly, preincubation with the GSH-depleting agents, l-buthionine-[S,R]-sulfoximine or diethyl maleate, prevented NO-mediated (59)Fe mobilization. This effect was reversed by incubating cells with N-acetyl-l-cysteine that reconstitutes GSH. These results indicate that GSH levels are essential for NO-mediated (59)Fe efflux. Hence, d-Glc metabolism via the hexose monophosphate shunt resulting in the generation of GSH may be essential for NO-mediated (59)Fe release. These results have important implications for intracellular signaling by NO and also NO-mediated cytotoxicity of activated macrophages that is due, in part, to iron release from tumor target cells.
Collapse
Affiliation(s)
- R N Watts
- Iron Metabolism and Chelation Group, the Heart Research Institute, 145 Missenden Rd, Camperdown, Sydney, New South Wales 2050, Australia
| | | |
Collapse
|
49
|
Ding H, Demple B. Direct nitric oxide signal transduction via nitrosylation of iron-sulfur centers in the SoxR transcription activator. Proc Natl Acad Sci U S A 2000; 97:5146-50. [PMID: 10805777 PMCID: PMC25796 DOI: 10.1073/pnas.97.10.5146] [Citation(s) in RCA: 236] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nitric oxide (NO) has diverse roles in intercellular communication and (at higher levels) in immune-mediated cell killing. NO reacts with many cellular targets, with cell-killing effects correlated to inactivation of key enzymes through nitrosylation of their iron-sulfur centers. SoxR protein, a redox-sensitive transcription activator dependent on the oxidation state of its binuclear iron-sulfur ([2Fe-2S]) centers, is also activated in Escherichia coli on exposure to macrophage-generated NO. We show here that SoxR activation by NO occurs through direct modification of the [2Fe-2S] centers to form protein-bound dinitrosyl-iron-dithiol adducts, which we have observed both in intact bacterial cells and in purified SoxR after NO treatment. Functional activation through nitrosylation of iron-sulfur centers contrasts with the inactivation typically caused by this modification. Purified, nitrosylated SoxR has transcriptional activity similar to that of oxidized SoxR and is relatively stable. In contrast, nitrosylated SoxR is short-lived in intact cells, indicative of mechanisms that actively dispose of nitrosylated iron-sulfur centers.
Collapse
Affiliation(s)
- H Ding
- Division of Toxicology, Department of Cancer Cell Biology, Harvard School of Public Health, Boston, MA 02115, USA
| | | |
Collapse
|
50
|
Bustamante J, Bersier G, Romero M, Badin RA, Boveris A. Nitric oxide production and mitochondrial dysfunction during rat thymocyte apoptosis. Arch Biochem Biophys 2000; 376:239-47. [PMID: 10775408 DOI: 10.1006/abbi.2000.1716] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Production of nitric oxide (NO) by mitochondrial membranes as methemoglobin formation sensitive to N(G)-methyl-l-arginine inhibition and mitochondrial O(2) consumption in metabolic states 3 and 4 and the respiratory control (state 3/state 4) were measured at early stages of rat thymocyte apoptosis. Programmed cell death was induced by addition of methylprednisolone and etoposide to thymocyte suspensions. Increased NO production by mitochondrial membranes was observed after 30 min of methylprednisolone and etoposide addition and was accompanied by mitochondrial respiratory impairment as an early phenomenon in apoptotic thymocytes. The respiratory control in isolated mitochondria from untreated thymocytes was 4.2 +/- 0.2 and decreased to 3.1 +/- 0.2 and 1.9 +/- 0.3 after 1 h of methylprednisolone and etoposide treatment, respectively. The low mitochondrial respiratory control was accompanied by a marked decrease in GSH and cytochrome c content. Moreover, an inhibitory effect in the amount of apoptosis due to thymocyte pretreatment with N(G)-methyl-l-arginine and N(omega)-nitro-(l)-arginine (l-NNA), indicate that nitric oxide production is closely involved in the signaling of rat thymocyte apoptosis.
Collapse
Affiliation(s)
- J Bustamante
- Laboratory of Free Radical Biology, School of Pharmacy and Biochemistry, University of Buenos Aires, Junin 956, Buenos Aires, 1113, Argentina.
| | | | | | | | | |
Collapse
|