1
|
Guinard P, Hostachy S, Diebold L, Pécaut J, Le Goff A, Duboc C, Delangle P. Outstanding Superoxide Dismutase Catalytic Activity Of Simple Peptide-Based Nickel(II) Complexes. Angew Chem Int Ed Engl 2024; 63:e202409343. [PMID: 39012328 DOI: 10.1002/anie.202409343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/17/2024]
Abstract
We present here the most active synthetic Ni superoxide dismutase (NiSOD) mimic reported to date. Reactive oxygen species are aggressive compounds, which concentrations are tightly regulated in vivo. Among them, the superoxide anion, O2⋅-, is controlled by superoxide dismutases. Capitalizing on the versatility of the Amino-Terminal CuII- and NiII-binding (ATCUN) peptide motif, we introduced positive charges around the NiII center to favor the interaction with the superoxide radical anion. At physiological pH, the pentapeptide H-Cys-His-Cys-Arg-Arg-NH2 coordinates NiII after the deprotonation of one thiol, two amides, and either the second thiol or the N-terminal ammonium, leading to an equilibrium between the two N3S1 and N2S2 coordination modes. Under catalytic conditions, a kcat value of 8.6(4)×106 L.mol-1.s-1 was measured. Within the first second, the catalyst remained undegraded with quantitative consumption of O2⋅- (completed up to 37 catalytic cycles). An extra arginine (Arg) was introduced at the peptide C-terminus to increase the global charge of the NiII complex from +1 to +2. This had no effect on the catalytic performance, highlighting the critical role of charge distribution in space as a determining factor influencing the reactivity.
Collapse
Affiliation(s)
- Pawel Guinard
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Sarah Hostachy
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Léa Diebold
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Jacques Pécaut
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| | - Alan Le Goff
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Carole Duboc
- Univ. Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Pascale Delangle
- Univ. Grenoble Alpes, CEA, CNRS, Grenoble INP, IRIG, SyMMES, 38000, Grenoble, France
| |
Collapse
|
2
|
Richaud A, Méndez F, Barba-Behrens N, Florian P, Medina-Campos ON, Pedraza-Chaverri J. Electrophilic Modulation of the Superoxide Anion Radical Scavenging Ability of Copper(II) Complexes with 4-Methyl Imidazole. J Phys Chem A 2021; 125:2394-2401. [PMID: 33754722 DOI: 10.1021/acs.jpca.0c10654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Three Cu(II) coordination compounds with 4-methyl imidazole were obtained, such as [Cu(C4H6N2)4(NO3)2], [Cu(C4H6N2)4Br2], and [Cu(C4H6N2)4Cl2]. Crystallographic studies confirmed their structural similarity with Cu(II) in the active site of endogenous copper-zinc superoxide dismutase (CuZn-SOD). The superoxide anion radical (O2•-) scavenging activity was evaluated by the non-enzymatic experimental assay and followed the trend [Cu(C4H6N2)4(NO3)2] > [Cu(C4H6N2)4Br2] > [Cu(C4H6N2)4Cl2]. The density functional theory and the hard and soft acids and bases principle showed the importance of the electron-deficient character of Cu(II) in the chemical reactivity of the coordination compounds; Cu(II) is the softest site in the molecule and it is preferred for the nucleophilic and radical attacks of the soft O2•-. A simple rule was obtained: "the electron-deficient character of Cu(II) is the key index for the O2•- scavenging activity and is modulated by the electron-releasing counteranion effect on the coordination compound".
Collapse
Affiliation(s)
- Arlette Richaud
- Departamento de Química, División de Ciencias Básicas e Ingeniería, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-534, Ciudad de México 09340, México.,Departamento de Química Inorgánica Facultad de Química, Universidad Nacional. Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México.,Le Studium Research Fellow, Loire Valley Institute for Advanced Studies, Orléans & Tours, France.,CEMHTI-CNRS, 1 Avenue de La Recherche Scientifique, Orléans 45100, France
| | - Francisco Méndez
- Departamento de Química, División de Ciencias Básicas e Ingeniería, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-534, Ciudad de México 09340, México.,Le Studium Research Fellow, Loire Valley Institute for Advanced Studies, Orléans & Tours, France.,CEMHTI-CNRS, 1 Avenue de La Recherche Scientifique, Orléans 45100, France
| | - Noráh Barba-Behrens
- Departamento de Química Inorgánica Facultad de Química, Universidad Nacional. Autónoma de México, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, México
| | - Pierre Florian
- CEMHTI-CNRS, 1 Avenue de La Recherche Scientifique, Orléans 45100, France
| | - Omar N Medina-Campos
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, México, D.F. 04510, México
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, México, D.F. 04510, México
| |
Collapse
|
3
|
Pinto VHA, Falcão NKSM, Mariz-Silva B, Fonseca MG, Rebouças JS. Robust Mn(III) N-pyridylporphyrin-based biomimetic catalysts for hydrocarbon oxidations: heterogenization on non-functionalized silica gel versus chloropropyl-functionalized silica gel. Dalton Trans 2020; 49:16404-16418. [PMID: 32633298 DOI: 10.1039/d0dt01383h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Two classes of heterogenized biomimetic catalysts were prepared and characterized for hydrocarbon oxidations: (1) by covalent anchorage of the three Mn(iii) meso-tetrakis(2-, 3-, or 4-pyridyl)porphyrin isomers by in situ alkylation with chloropropyl-functionalized silica gel (Sil-Cl) to yield Sil-Cl/MnPY (Y = 1, 2, 3) materials, and (2) by electrostatic immobilization of the three Mn(iii) meso-tetrakis(N-methylpyridinium-2, 3, or 4-yl)porphyrin isomers (MnPY, Y = 4, 5, 6) on non-modified silica gel (SiO2) to yield SiO2/MnPY (Y = 4, 5, 6) materials. Silica gel used was of column chromatography grade and Mn porphyrin loadings were deliberately kept at a low level (0.3% w/w). These resulting materials were explored as catalysts for iodosylbenzene (PhIO) oxidation of cyclohexane, n-heptane, and adamantane to yield the corresponding alcohols and ketones; the oxidation of cyclohexanol to cyclohexanone was also investigated. The heterogenized catalysts exhibited higher efficiency and selectivity than the corresponding Mn porphyrins under homogeneous conditions. Recycling studies were consistent with low leaching/destruction of the supported Mn porphyrins. The Sil-Cl/MnPY catalysts were more efficient and more selective than SiO2/MnPY ones; alcohol selectivity may be associated with hydrophobic silica surface modification reminiscent of biological cytochrome P450 oxidations. The use of widespread, column chromatography, amorphous silica yielded Sil-Cl/MnPY or SiO2/MnPY catalysts considerably more efficient than the corresponding, previously reported materials with mesoporous Santa Barbara Amorphous No 15 (SBA-15) silica. Among the materials studied, in situ derivatization of Mn(iii) 2-N-pyridylporphyrin by covalent immobilization on Sil-Cl to yield Sil-Cl/MnP1 showed the best catalytic performance with high stability against oxidative destruction and reusability/recyclability.
Collapse
Affiliation(s)
- Victor Hugo A Pinto
- Department of Chemistry, CCEN, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil.
| | | | | | | | | |
Collapse
|
4
|
Stojičkov M, Sturm S, Čobeljić B, Pevec A, Jevtović M, Scheitler A, Radanović D, Senft L, Turel I, Andjelković K, Miehlich M, Meyer K, Ivanović‐Burmazović I. Cobalt(II), Zinc(II), Iron(III), and Copper(II) Complexes Bearing Positively Charged Quaternary Ammonium Functionalities: Synthesis, Characterization, Electrochemical Behavior, and SOD Activity. Eur J Inorg Chem 2020. [DOI: 10.1002/ejic.202000415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Marko Stojičkov
- Faculty of Chemistry University of Belgrade Studentski trg 12‐16 11000 Belgrade Serbia
| | - Sabrina Sturm
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
| | - Božidar Čobeljić
- Faculty of Chemistry University of Belgrade Studentski trg 12‐16 11000 Belgrade Serbia
| | - Andrej Pevec
- Faculty of Chemistry and Chemical Technology University of Ljubljana Večna pot 113 1000 Ljubljana Slovenia
| | - Mima Jevtović
- Faculty of Chemistry University of Belgrade Studentski trg 12‐16 11000 Belgrade Serbia
| | - Andreas Scheitler
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
| | - Dušanka Radanović
- Institute of Chemistry Technology and Metallurgy University of Belgrade Njegoševa 12, P.O. Box 815 11000 Belgrade Serbia
| | - Laura Senft
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology University of Ljubljana Večna pot 113 1000 Ljubljana Slovenia
| | - Katarina Andjelković
- Faculty of Chemistry University of Belgrade Studentski trg 12‐16 11000 Belgrade Serbia
| | - Matthias Miehlich
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
| | - Karsten Meyer
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
| | - Ivana Ivanović‐Burmazović
- Department of Chemistry and Pharmacy Friedrich‐Alexander University Erlangen‐Nürnberg Egerlandstr. 1 91058 Erlangen Germany
- Department Chemie Ludwigs‐Maximilians‐Universität Butenandtstraße 5‐13 81377 München Germany
| |
Collapse
|
5
|
Mathieu E, Bernard AS, Ching HYV, Somogyi A, Medjoubi K, Fores JR, Bertrand HC, Vincent A, Trépout S, Guerquin-Kern JL, Scheitler A, Ivanović-Burmazović I, Seksik P, Delsuc N, Policar C. Anti-inflammatory activity of superoxide dismutase mimics functionalized with cell-penetrating peptides. Dalton Trans 2020; 49:2323-2330. [DOI: 10.1039/c9dt04619d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A superoxide dismutase mimic was functionalized with three peptides: -R9, -RRWWRRWRR or -Fx-r-Fx-K (MPP). They were studied in intestinal epithelial cells in an inorganic cellular chemistry approach: quantification, distribution and bio-activity.
Collapse
|
6
|
Franke A, Scheitler A, Kenkel I, Lippert R, Zahl A, Balbinot D, Jux N, Ivanović-Burmazović I. Positive Charge on Porphyrin Ligand and Nature of Metal Center Define Basic Physicochemical Properties of Cationic Manganese and Iron Porphyrins in Aqueous Solution. Inorg Chem 2019; 58:9618-9630. [DOI: 10.1021/acs.inorgchem.8b03381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Alicja Franke
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Andreas Scheitler
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Isabell Kenkel
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Rainer Lippert
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Achim Zahl
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Domenico Balbinot
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | - Norbert Jux
- Department of Chemistry and Pharmacy, University Erlangen−Nuremberg, 91058 Erlangen, Germany
| | | |
Collapse
|
7
|
|
8
|
Ching HV, Kenkel I, Delsuc N, Mathieu E, Ivanović-Burmazović I, Policar C. Bioinspired superoxide-dismutase mimics: The effects of functionalization with cationic polyarginine peptides. J Inorg Biochem 2016; 160:172-9. [DOI: 10.1016/j.jinorgbio.2016.01.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/27/2015] [Accepted: 01/25/2016] [Indexed: 10/22/2022]
|
9
|
Leung CH, Lin S, Zhong HJ, Ma DL. Metal complexes as potential modulators of inflammatory and autoimmune responses. Chem Sci 2015; 6:871-884. [PMID: 28660015 PMCID: PMC5472922 DOI: 10.1039/c4sc03094j] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/07/2014] [Indexed: 01/05/2023] Open
Abstract
Over the past few decades, the realm of inorganic medicinal chemistry has been dominated by the study of the anti-cancer properties of transition metal complexes, particularly those based on platinum or ruthenium. However, comparatively less attention has been focused on the development of metal complexes for the treatment of inflammatory or autoimmune diseases. Metal complexes possess a number of advantages that render them as attractive alternatives to organic small molecules for the development of therapeutic agents. In this perspective, we highlight recent examples in the development of transition metal complexes as modulators of inflammatory and autoimmune responses. The studies presented here serve to highlight the potential of transition metal complexes in modulating inflammatory or immune pathways in cells.
Collapse
Affiliation(s)
- Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences , University of Macau , Macao , China .
| | - Sheng Lin
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China .
| | - Hai-Jing Zhong
- State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences , University of Macau , Macao , China .
| | - Dik-Lung Ma
- Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong , China .
| |
Collapse
|
10
|
Haber A, Gross Z. Catalytic antioxidant therapy by metallodrugs: lessons from metallocorroles. Chem Commun (Camb) 2015; 51:5812-27. [DOI: 10.1039/c4cc08715a] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This article provides a perspective on the utility of metal-based catalytic antioxidants for disease prevention or treatment, with focus on their mode of action and its dependence (DCA) or independence (ICA) on the involvement of cofactors.
Collapse
Affiliation(s)
- Adi Haber
- Schulich Faculty of Chemistry
- Technion – Israel Institute of Technology
- Technion City
- Israel
| | - Zeev Gross
- Schulich Faculty of Chemistry
- Technion – Israel Institute of Technology
- Technion City
- Israel
| |
Collapse
|
11
|
Tovmasyan A, Reboucas JS, Benov L. Simple biological systems for assessing the activity of superoxide dismutase mimics. Antioxid Redox Signal 2014; 20:2416-36. [PMID: 23964890 PMCID: PMC4005499 DOI: 10.1089/ars.2013.5576] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Half a century of research provided unambiguous proof that superoxide and species derived from it-reactive oxygen species (ROS)-play a central role in many diseases and degenerative processes. This stimulated the search for pharmaceutical agents that are capable of preventing oxidative damage, and methods of assessing their therapeutic potential. RECENT ADVANCES The limitations of superoxide dismutase (SOD) as a therapeutic tool directed attention to small molecules, SOD mimics, that are capable of catalytically scavenging superoxide. Several groups of compounds, based on either metal complexes, including metalloporphyrins, metallocorroles, Mn(II) cyclic polyamines, and Mn(III) salen derivatives, or non-metal based compounds, such as fullerenes, nitrones, and nitroxides, have been developed and studied in vitro and in vivo. Very few entered clinical trials. CRITICAL ISSUES AND FUTURE DIRECTIONS Development of SOD mimics requires in-depth understanding of their mechanisms of biological action. Elucidation of both molecular features, essential for efficient ROS-scavenging in vivo, and factors limiting the potential side effects requires biologically relevant and, at the same time, relatively simple testing systems. This review discuses the advantages and limitations of genetically engineered SOD-deficient unicellular organisms, Escherichia coli and Saccharomyces cerevisiae as tools for investigating the efficacy and mechanisms of biological actions of SOD mimics. These simple systems allow the scrutiny of the minimal requirements for a functional SOD mimic: the association of a high catalytic activity for superoxide dismutation, low toxicity, and an efficient cellular uptake/biodistribution.
Collapse
Affiliation(s)
- Artak Tovmasyan
- 1 Department of Radiation Oncology, Duke University Medical Center , Durham, North Carolina
| | | | | |
Collapse
|
12
|
Batinic-Haberle I, Tovmasyan A, Roberts ERH, Vujaskovic Z, Leong KW, Spasojevic I. SOD therapeutics: latest insights into their structure-activity relationships and impact on the cellular redox-based signaling pathways. Antioxid Redox Signal 2014; 20:2372-415. [PMID: 23875805 PMCID: PMC4005498 DOI: 10.1089/ars.2012.5147] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 06/30/2013] [Accepted: 07/22/2013] [Indexed: 01/23/2023]
Abstract
SIGNIFICANCE Superoxide dismutase (SOD) enzymes are indispensable and ubiquitous antioxidant defenses maintaining the steady-state levels of O2·(-); no wonder, thus, that their mimics are remarkably efficacious in essentially any animal model of oxidative stress injuries thus far explored. RECENT ADVANCES Structure-activity relationship (half-wave reduction potential [E1/2] versus log kcat), originally reported for Mn porphyrins (MnPs), is valid for any other class of SOD mimics, as it is dominated by the superoxide reduction and oxidation potential. The biocompatible E1/2 of ∼+300 mV versus normal hydrogen electrode (NHE) allows powerful SOD mimics as mild oxidants and antioxidants (alike O2·(-)) to readily traffic electrons among reactive species and signaling proteins, serving as fine mediators of redox-based signaling pathways. Based on similar thermodynamics, both SOD enzymes and their mimics undergo similar reactions, however, due to vastly different sterics, with different rate constants. CRITICAL ISSUES Although log kcat(O2·(-)) is a good measure of therapeutic potential of SOD mimics, discussions of their in vivo mechanisms of actions remain mostly of speculative character. Most recently, the therapeutic and mechanistic relevance of oxidation of ascorbate and glutathionylation and oxidation of protein thiols by MnP-based SOD mimics and subsequent inactivation of nuclear factor κB has been substantiated in rescuing normal and killing cancer cells. Interaction of MnPs with thiols seems to be, at least in part, involved in up-regulation of endogenous antioxidative defenses, leading to the healing of diseased cells. FUTURE DIRECTIONS Mechanistic explorations of single and combined therapeutic strategies, along with studies of bioavailability and translational aspects, will comprise future work in optimizing redox-active drugs.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Emily R. H. Roberts
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- King Abdulaziz University, Jeddah, Saudi Arabia Kingdom
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical School, Durham, North Carolina
| |
Collapse
|
13
|
Weitner T, Kos I, Sheng H, Tovmasyan A, Reboucas JS, Fan P, Warner DS, Vujaskovic Z, Batinic-Haberle I, Spasojevic I. Comprehensive pharmacokinetic studies and oral bioavailability of two Mn porphyrin-based SOD mimics, MnTE-2-PyP5+ and MnTnHex-2-PyP5+. Free Radic Biol Med 2013; 58:73-80. [PMID: 23328731 PMCID: PMC3763724 DOI: 10.1016/j.freeradbiomed.2013.01.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 12/20/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
The cationic, ortho Mn(III) N-alkylpyridylporphyrins (alkyl=ethyl, E, and n-hexyl, nHex) MnTE-2-PyP(5+) (AEOL10113, FBC-007) and MnTnHex-2-PyP(5+) have proven efficacious in numerous in vivo animal models of diseases having oxidative stress in common. The remarkable therapeutic efficacy observed is due to their: (1) ability to catalytically remove O2(•-) and ONOO(-) and other reactive species; (2) ability to modulate redox-based signaling pathways; (3) accumulation within critical cellular compartments, i.e., mitochondria; and (4) ability to cross the blood-brain barrier. The similar redox activities of both compounds are related to the similar electronic and electrostatic environments around the metal active sites, whereas their different bioavailabilities are presumably influenced by the differences in lipophilicity, bulkiness, and shape. Both porphyrins are water soluble, but MnTnHex-2-PyP(5+) is approximately 4 orders of magnitude more lipophilic than MnTE-2-PyP(5+), which should positively affect its ability to pass through biological membranes, making it more efficacious in vivo at lower doses. To gain insight into the in vivo tissue distribution of Mn porphyrins and its impact upon their therapeutic efficacy and mechanistic aspects of action, as well as to provide data that would ensure proper dosing regimens, we conducted comprehensive pharmacokinetic (PK) studies for 24h after single-dose drug administration. The porphyrins were administered intravenously (iv), intraperitoneally (ip), and via oral gavage at the following doses: 10mg/kg MnTE-2-PyP(5+) and 0.5 or 2mg/kg MnTnHex-2-PyP(5+). Drug levels in plasma and various organs (liver, kidney, spleen, heart, lung, brain) were determined and PK parameters calculated (Cmax, C24h, tmax, and AUC). Regardless of high water solubility and pentacationic charge of these Mn porphyrins, they are orally available. The oral availability (based on plasma AUCoral/AUCiv) is 23% for MnTE-2-PyP(5+) and 21% for MnTnHex-2-PyP(5+). Despite the fivefold lower dose administered, the AUC values for liver, heart, and spleen are higher for MnTnHex-2-PyP(5+) than for MnTE-2-PyP(5+) (and comparable for other organs), clearly demonstrating the better tissue penetration and tissue retention of the more lipophilic MnTnHex-2-PyP(5+).
Collapse
Affiliation(s)
- Tin Weitner
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ivan Kos
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Julio S. Reboucas
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ping Fan
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - David S. Warner
- Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
14
|
Tovmasyan A, Sheng H, Weitner T, Arulpragasam A, Lu M, Warner DS, Vujaskovic Z, Spasojevic I, Batinic-Haberle I. Design, mechanism of action, bioavailability and therapeutic effects of mn porphyrin-based redox modulators. Med Princ Pract 2012; 22:103-30. [PMID: 23075911 PMCID: PMC3640855 DOI: 10.1159/000341715] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 07/01/2012] [Indexed: 12/18/2022] Open
Abstract
Based on aqueous redox chemistry and simple in vivo models of oxidative stress, Escherichia coli and Saccharomyces cerevisiae, the cationic Mn(III) N-substituted pyridylporphyrins (MnPs) have been identified as the most potent cellular redox modulators within the porphyrin class of drugs; their efficacy in animal models of diseases that have oxidative stress in common is based on their high ability to catalytically remove superoxide, peroxynitrite, carbonate anion radical, hypochlorite, nitric oxide, lipid peroxyl and alkoxyl radicals, thus suppressing the primary oxidative event. While doing so MnPs could couple with cellular reductants and redox-active proteins. Reactive species are widely accepted as regulators of cellular transcriptional activity: minute, nanomolar levels are essential for normal cell function, while submicromolar or micromolar levels impose oxidative stress, which is evidenced in increased inflammatory and immune responses. By removing reactive species, MnPs affect redox-based cellular transcriptional activity and consequently secondary oxidative stress, and in turn inflammatory processes. The equal ability to reduce and oxidize superoxide during the dismutation process and recently accumulated results suggest that pro-oxidative actions of MnPs may also contribute to their therapeutic effects. All our data identify the superoxide dismutase-like activity, estimated by log k(cat)O2-*), as a good measure for the therapeutic efficacy of MnPs. Their accumulation in mitochondria and their ability to cross the blood-brain barrier contribute to their remarkable efficacy. We summarize herein the therapeutic effects of MnPs in cancer, central nervous system injuries, diabetes, their radioprotective action and potential for imaging. Few of the most potent modulators of cellular redox-based pathways, MnTE2-PyP5+, MnTDE-2-ImP5+, MnTnHex-2-PyP5+ and MnTnBuOE-2-PyP5+, are under preclinical and clinical development.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Tin Weitner
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Amanda Arulpragasam
- Department of Duke University Neuroscience Undergraduate
Program, Duke University Medical Center, Durham, N.C., USA
| | - Miaomiao Lu
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
- Department of Department of Anesthesiology, Second Affiliated
Hospital, Zhengzhou University, Zhengzhou, China
| | - David S. Warner
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham,
N.C., USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| |
Collapse
|
15
|
Kalmár J, Biri B, Lente G, Bányai I, Budimir A, Biruš M, Batinić-Haberle I, Fábián I. Detailed mechanism of the autoxidation of N-hydroxyurea catalyzed by a superoxide dismutase mimic Mn(III) porphyrin: formation of the nitrosylated Mn(II) porphyrin as an intermediate. Dalton Trans 2012; 41:11875-84. [PMID: 22911446 DOI: 10.1039/c2dt31200j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The in vitro autoxidation of N-hydroxyurea (HU) is catalyzed by Mn(III)TTEG-2-PyP(5+), a synthetic water soluble Mn(III) porphyrin which is also a potent mimic of the enzyme superoxide dismutase. The detailed mechanism of the reaction is deduced from kinetic studies under basic conditions mostly based on data measured at pH = 11.7 but also including some pH-dependent observations in the pH range 9-13. The major intermediates were identified by UV-vis spectroscopy and electrospray ionization mass spectrometry. The reaction starts with a fast axial coordination of HU to the metal center of Mn(III)TTEG-2-PyP(5+), which is followed by a ligand-to-metal electron transfer to get Mn(II)TTEG-2-PyP(4+) and the free radical derived from HU (HU˙). Nitric oxide (NO) and nitroxyl (HNO) are minor intermediates. The major pathway for the formation of the most significant intermediate, the {MnNO} complex of Mn(II)TTEG-2-PyP(4+), is the reaction of Mn(II)TTEG-2-PyP(4+) with NO. We have confirmed that the autoxidation of the intermediates opens alternative reaction channels, and the process finally yields NO(2)(-) and the initial Mn(III)TTEG-2-PyP(5+). The photochemical release of NO from the {MnNO} intermediate was also studied. Kinetic simulations were performed to validate the deduced rate constants. The investigated reaction has medical implications: the accelerated production of NO and HNO from HU may be utilized for therapeutic purposes.
Collapse
Affiliation(s)
- József Kalmár
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen, Hungary H-4010, POB-21
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Pinto VHA, Carvalhoda-Silva D, Santos JLMS, Weitner T, Fonseca MG, Yoshida MI, Idemori YM, Batinić-Haberle I, Rebouças JS. Thermal stability of the prototypical Mn porphyrin-based superoxide dismutase mimic and potent oxidative-stress redox modulator Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin chloride, MnTE-2-PyP(5+). J Pharm Biomed Anal 2012; 73:29-34. [PMID: 22503130 DOI: 10.1016/j.jpba.2012.03.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 03/14/2012] [Accepted: 03/16/2012] [Indexed: 10/28/2022]
Abstract
Cationic Mn porphyrins are among the most potent catalytic antioxidants and/or cellular redox modulators. Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin chloride (MnTE-2-PyPCl(5)) is the Mn porphyrin most studied in vivo and has successfully rescued animal models of a variety of oxidative stress-related diseases. The stability of an authentic MnTE-2-PyPCl(5) sample was investigated hereon by thermogravimetric, derivative thermogravimetric, and differential thermal analyses (TG/DTG/DTA), under dynamic air, followed by studies at selected temperatures to evaluate the decomposition path and appropriate conditions for storage and handling of these materials. All residues were analyzed by thin-layer chromatography (TLC) and UV-vis spectroscopy. Three thermal processes were observed by TG/DTG. The first event (endothermic) corresponded to dehydration, and did not alter the MnTE-2-PyPCl(5) moiety. The second event (endothermic) corresponded to the loss of EtCl (dealkylation), which was characterized by gas chromatography-mass spectrometry. The residue at 279°C had UV-vis and TLC data consistent with those of the authentic, completely dealkylated analog, MnT-2-PyPCl. The final, multi-step event corresponded to the loss of the remaining organic matter to yield Mn(3)O(4) which was characterized by IR spectroscopy. Isothermal treatment at 188°C under static air for 3h yielded a mixture of partially dealkylated MnPs and traces of the free-base, dealkylated ligand, H(2)T-2-PyP, which reveals that dealkylation is accompanied by thermal demetallation under static air conditions. Dealkylation was not observed if the sample was heated as a solid or in aqueous solution up to ∼100°C. Whereas moderate heating changes sample composition by loss of H(2)O, the dehydrated sample is indistinguishable from the original sample upon dissolution in water, which indicates that catalytic activity (on Mn basis) remains unaltered. Evidently, dealkylation at high temperature compromises sample activity.
Collapse
Affiliation(s)
- Victor H A Pinto
- Departamento de Química, CCEN, Universidade Federal da Paraíba, João Pessoa PB 58051-900, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Batinic-Haberle I, Rajic Z, Tovmasyan A, Ye X, Leong KW, Dewhirst MW, Vujaskovic Z, Benov L, Spasojevic I. Diverse functions of cationic Mn(III) N-substituted pyridylporphyrins, recognized as SOD mimics. Free Radic Biol Med 2011; 51:1035-53. [PMID: 21616142 PMCID: PMC3178885 DOI: 10.1016/j.freeradbiomed.2011.04.046] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/30/2011] [Accepted: 04/29/2011] [Indexed: 10/18/2022]
Abstract
Oxidative stress, a redox imbalance between the endogenous reactive species and antioxidant systems, is common to numerous pathological conditions such as cancer, central nervous system injuries, radiation injury, diabetes etc. Therefore, compounds able to reduce oxidative stress have been actively sought for over 3 decades. Superoxide is the major species involved in oxidative stress either in its own right or through its progeny, such as ONOO⁻, H₂O₂, •OH, CO₃•⁻, and •NO₂. Hence, the very first compounds developed in the late 1970-ies were the superoxide dismutase (SOD) mimics. Thus far the most potent mimics have been the cationic meso Mn(III) N-substituted pyridylporphyrins and N,N'-disubstituted imidazolylporphyrins (MnPs), some of them with k(cat)(O₂·⁻) similar to the k(cat) of SOD enzymes. Most frequently studied are ortho isomers MnTE-2-PyP⁵⁺, MnTnHex-2-PyP⁵⁺, and MnTDE-2-ImP⁵⁺. The ability to disproportionate O₂·⁻ parallels their ability to remove the other major oxidizing species, peroxynitrite, ONOO⁻. The same structural feature that gives rise to the high k(cat)(O₂·⁻) and k(red)(ONOO⁻), allows MnPs to strongly impact the activation of the redox-sensitive transcription factors, HIF-1α, NF-κB, AP-1, and SP-1, and therefore modify the excessive inflammatory and immune responses. Coupling with cellular reductants and other redox-active endogenous proteins seems to be involved in the actions of Mn porphyrins. While hydrophilic analogues, such as MnTE-2-PyP⁵⁺ and MnTDE-2-ImP⁵⁺ are potent in numerous animal models of diseases, the lipophilic analogues, such as MnTnHex-2-PyP⁵⁺, were developed to cross blood brain barrier and target central nervous system and critical cellular compartments, mitochondria. The modification of its structure, aimed to preserve the SOD-like potency and lipophilicity, and diminish the toxicity, has presently been pursued. The pulmonary radioprotection by MnTnHex-2-PyP⁵⁺ was the first efficacy study performed successfully with non-human primates. The Phase I toxicity clinical trials were done on amyotrophic lateral sclerosis patients with N,N'-diethylimidazolium analogue, MnTDE-2-ImP⁵⁺ (AEOL10150). Its aggressive development as a wide spectrum radioprotector by Aeolus Pharmaceuticals has been supported by USA Federal government. The latest generation of compounds, bearing oxygens in pyridyl substituents is presently under aggressive development for cancer and CNS injuries at Duke University and is supported by Duke Translational Research Institute, The Wallace H. Coulter Translational Partners Grant Program, Preston Robert Tisch Brain Tumor Center at Duke, and National Institute of Allergy and Infectious Diseases. Metal center of cationic MnPs easily accepts and donates electrons as exemplified in the catalysis of O₂·⁻ dismutation. Thus such compounds may be equally good anti- and pro-oxidants; in either case the beneficial therapeutic effects may be observed. Moreover, while the in vivo effects may appear antioxidative, the mechanism of action of MnPs that produced such effects may be pro-oxidative; the most obvious example being the inhibition of NF-κB. The experimental data therefore teach us that we need to distinguish between the mechanism/s of action/s of MnPs and the effects we observe. A number of factors impact the type of action of MnPs leading to favorable therapeutic effects: levels of reactive species and oxygen, levels of endogenous antioxidants (enzymes and low-molecular compounds), levels of MnPs, their site of accumulation, and the mutual encounters of all of those species. The complexity of in vivo redox systems and the complex redox chemistry of MnPs challenge and motivate us to further our understanding of the physiology of the normal and diseased cell with ultimate goal to successfully treat human diseases.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| | - Zrinka Rajic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaodong Ye
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait School of Medicine, Kuwait
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| |
Collapse
|
18
|
Iranzo O. Manganese complexes displaying superoxide dismutase activity: A balance between different factors. Bioorg Chem 2011; 39:73-87. [DOI: 10.1016/j.bioorg.2011.02.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/14/2011] [Accepted: 02/14/2011] [Indexed: 01/05/2023]
|
19
|
Batinić-Haberle I, Rebouças JS, Spasojević I. Superoxide dismutase mimics: chemistry, pharmacology, and therapeutic potential. Antioxid Redox Signal 2010; 13:877-918. [PMID: 20095865 PMCID: PMC2935339 DOI: 10.1089/ars.2009.2876] [Citation(s) in RCA: 390] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidative stress has become widely viewed as an underlying condition in a number of diseases, such as ischemia-reperfusion disorders, central nervous system disorders, cardiovascular conditions, cancer, and diabetes. Thus, natural and synthetic antioxidants have been actively sought. Superoxide dismutase is a first line of defense against oxidative stress under physiological and pathological conditions. Therefore, the development of therapeutics aimed at mimicking superoxide dismutase was a natural maneuver. Metalloporphyrins, as well as Mn cyclic polyamines, Mn salen derivatives and nitroxides were all originally developed as SOD mimics. The same thermodynamic and electrostatic properties that make them potent SOD mimics may allow them to reduce other reactive species such as peroxynitrite, peroxynitrite-derived CO(3)(*-), peroxyl radical, and less efficiently H(2)O(2). By doing so SOD mimics can decrease both primary and secondary oxidative events, the latter arising from the inhibition of cellular transcriptional activity. To better judge the therapeutic potential and the advantage of one over the other type of compound, comparative studies of different classes of drugs in the same cellular and/or animal models are needed. We here provide a comprehensive overview of the chemical properties and some in vivo effects observed with various classes of compounds with a special emphasis on porphyrin-based compounds.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
20
|
Fernandes AS, Gaspar J, Cabral MF, Rueff J, Castro M, Batinic-Haberle I, Costa J, Oliveira NG. Protective role of ortho-substituted Mn(III) N-alkylpyridylporphyrins against the oxidative injury induced by tert-butylhydroperoxide. Free Radic Res 2010; 44:430-40. [PMID: 20102317 DOI: 10.3109/10715760903555844] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The present work addresses the role of two ortho-substituted Mn(III) N-alkylpyridylporphyrins, alkyl being ethyl in MnTE-2-PyP(5+) and n-hexyl in MnTnHex-2-PyP(5+), on the protection against the oxidant tert-butylhydroperoxide (TBHP). Their protective role was studied in V79 cells using endpoints of cell viability (MTT and crystal violet assays), intracellular O(2)*- generation (dihydroethidium assay) and glutathione status (DTNB and monochlorobimane assays). MnPs per se did not show cytotoxicity (up to 25 microM, 24 h). The exposure to TBHP resulted in a significant decrease in cell viability and in an increase in the intracellular O(2)(*-) levels. Also, TBHP depleted total and reduced glutathione and increased GSSG. The two MnPs counteracted remarkably the effects of TBHP. Even at low concentrations, both MnPs were protective in terms of cell viability and abrogated the intracellular O(2)(*-) increase in a significant way. Also, they augmented markedly the total and reduced glutathione contents in TBHP-treated cells, highlighting the multiple mechanisms of protection of these SOD mimics, which at least in part may be ascribed to their electron-donating ability.
Collapse
Affiliation(s)
- Ana S Fernandes
- iMed.UL, Faculty of Pharmacy, University of Lisbon, Lisboa, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Budimir A, Kalmár J, Fábián I, Lente G, Bányai I, Batinić-Haberle I, Birus M. Water exchange rates of water-soluble manganese(III) porphyrins of therapeutical potential. Dalton Trans 2010; 39:4405-10. [PMID: 20422097 DOI: 10.1039/b926522h] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The activation parameters and the rate constants of the water-exchange reactions of Mn(III)TE-2-PyP(5+) (meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin) as cationic, Mn(III)TnHex-2-PyP(5+) (meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin) as sterically shielded cationic, and Mn(III)TSPP(3-) (meso-tetrakis(4-sulfonatophenyl)porphyrin) as anionic manganese(iii) porphyrins were determined from the temperature dependence of (17)O NMR relaxation rates. The rate constants at 298 K were obtained as 4.12 x 10(6) s(-1), 5.73 x 10(6) s(-1), and 2.74 x 10(7) s(-1), respectively. On the basis of the determined entropies of activation, an interchange-dissociative mechanism (I(d)) was proposed for the cationic complexes (DeltaS(double dagger) = approximately 0 J mol(-1) K(-1)) whereas a limiting dissociative mechanism (D) was proposed for Mn(III)TSPP(3-) complex (DeltaS(double dagger) = +79 J mol(-1) K(-1)). The obtained water exchange rate of Mn(III)TSPP(3-) corresponded well to the previously assumed value used by Koenig et al. (S. H. Koenig, R. D. Brown and M. Spiller, Magn. Reson. Med., 1987, 4, 52-260) to simulate the (1)H NMRD curves, therefore the measured value supports the theory developed for explaining the anomalous relaxivity of Mn(III)TSPP(3-) complex. A magnitude of the obtained water-exchange rate constants further confirms the suggested inner sphere electron transfer mechanism for the reactions of the two positively charged Mn(iii) porphyrins with the various biologically important oxygen and nitrogen reactive species. Due to the high biological and clinical relevance of the reactions that occur at the metal site of the studied Mn(iii) porphyrins, the determination of water exchange rates advanced our insight into their efficacy and mechanism of action, and in turn should impact their further development for both diagnostic (imaging) and therapeutic purposes.
Collapse
Affiliation(s)
- Ana Budimir
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovacića 1, 10000, Zagreb, Croatia
| | | | | | | | | | | | | |
Collapse
|
22
|
Kos I, Benov L, Spasojević I, Rebouças JS, Batinić-Haberle I. High lipophilicity of meta Mn(III) N-alkylpyridylporphyrin-based superoxide dismutase mimics compensates for their lower antioxidant potency and makes them as effective as ortho analogues in protecting superoxide dismutase-deficient Escherichia coli. J Med Chem 2009; 52:7868-72. [PMID: 19954250 DOI: 10.1021/jm900576g] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lipophilicity/bioavailibility of Mn(III) N-alkylpyridylporphyrin-based superoxide dismutase (SOD) mimics has a major impact on their in vivo ability to suppress oxidative stress. Meta isomers are less potent SOD mimics than ortho analogues but are 10-fold more lipophilic and more planar. Enhanced lipophilicity contributes to their higher accumulation in cytosol of SOD-deficient Escherichia coli, compensating for their lower potency; consequently, both isomers exert similar-to-identical protection of SOD-deficient E. coli. Thus meta isomers may be prospective therapeutics as are ortho porphyrins.
Collapse
Affiliation(s)
- Ivan Kos
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
23
|
Tabares LC, Gätjens J, Un S. Understanding the influence of the protein environment on the Mn(II) centers in Superoxide Dismutases using High-Field Electron Paramagnetic Resonance. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2009; 1804:308-17. [PMID: 19818880 DOI: 10.1016/j.bbapap.2009.09.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/16/2009] [Accepted: 09/23/2009] [Indexed: 01/20/2023]
Abstract
One of the most puzzling questions of manganese and iron superoxide dismutases (SODs) is what is the basis for their metal-specificity. This review summarizes our findings on the Mn(II) electronic structure of SODs and related synthetic models using high-field high-frequency electron paramagnetic resonance (HFEPR), a technique that is able to achieve a very detailed and quantitative information about the electronic structure of the Mn(II) ions. We have used HFEPR to compare eight different SODs, including iron, manganese and cambialistic proteins. This comparative approach has shown that in spite of their high structural homology each of these groups have specific spectroscopic and biochemical characteristics. This has allowed us to develop a model about how protein and metal interactions influence protein pK, inhibitor binding and the electronic structure of the manganese center. To better appreciate the thermodynamic prerequisites required for metal discriminatory SOD activity and their relationship to HFEPR spectroscopy, we review the work on synthetic model systems that functionally mimic Mn-and FeSOD. Using a single ligand framework, it was possible to obtain metal-discriminatory "activity" as well as variations in the HFEPR spectra that parallel those found in the proteins. Our results give new insights into protein-metal interactions from the perspective of the Mn(II) and new steps towards solving the puzzle of metal-specificity in SODs.
Collapse
Affiliation(s)
- Leandro C Tabares
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | | | | |
Collapse
|
24
|
Rabbani ZN, Spasojevic I, Zhang X, Moeller BJ, Haberle S, Vasquez-Vivar J, Dewhirst MW, Vujaskovic Z, Batinic-Haberle I. Antiangiogenic action of redox-modulating Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+), via suppression of oxidative stress in a mouse model of breast tumor. Free Radic Biol Med 2009; 47:992-1004. [PMID: 19591920 PMCID: PMC2749298 DOI: 10.1016/j.freeradbiomed.2009.07.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 06/13/2009] [Accepted: 07/02/2009] [Indexed: 12/20/2022]
Abstract
MnTE-2-PyP(5+) is a potent catalytic scavenger of reactive oxygen and nitrogen species, primarily superoxide and peroxynitrite. It therefore not only attenuates primary oxidative damage, but was found to modulate redox-based signaling pathways (HIF-1alpha, NF-kappaB, SP-1, and AP-1) and thus, in turn, secondary oxidative injury also. Cancer has been widely considered an oxidative stress condition. The goal of this study was to prove if and why a catalytic SOD mimic/peroxynitrite scavenger would exert anti-cancer effects, i.e., to evaluate whether the attenuation of the oxidative stress by MnTE-2-PyP(5+) could suppress tumor growth in a 4T1 mouse breast tumor model. Tumor cells were implanted into Balb/C mouse flanks. Three groups of mice (n=25) were studied: control (PBS) and 2 and 15 mg/kg/day of MnTE-2-PyP(5+) given subcutaneously twice daily starting when the tumors averaged 200 mm(3) (until they reached approximately 5-fold the initial volume). Intratumoral hypoxia (pimonidazole, carbonic anhydrase), HIF-1alpha, VEGF, proliferating capillary index (CD105), microvessel density (CD31), protein nitration, DNA oxidation (8-OHdG), NADPH oxidase (Nox-4), apoptosis (CD31), macrophage infiltration (CD68), and tumor drug levels were assessed. With 2 mg/kg/day a trend toward tumor growth delay was observed, and a significant trend was observed with 15 mg/kg/day. The 7.5-fold increase in drug dose was accompanied by a similar (6-fold) increase in tumor drug levels. Oxidative stress was largely attenuated as observed through the decreased levels of DNA damage, protein 3-nitrotyrosine, macrophage infiltration, and NADPH oxidase. Further, hypoxia was significantly decreased as were the levels of HIF-1alpha and VEGF. Consequently, suppression of angiogenesis was observed; both the microvessel density and the endothelial cell proliferation were markedly decreased. Our study indicates for the first time that MnTE-2-PyP(5+) has anti-cancer activity in its own right. The anti-cancer activity via HIF/VEGF pathways probably arises from the impact of the drug on the oxidative stress. Therefore, the catalytic scavenging of ROS/RNS by antioxidants, which in turn suppresses cellular transcriptional activity, could be an appropriate strategy for anti-cancer therapy. Enhancement of the anti-cancer effects may be achieved by optimizing the dosing regime, utilizing more bioavailable Mn porphyrins (MnP), and combining MnP treatment with irradiation, hyperthermia, and chemotherapy. Mn porphyrins may be advantageous compared to other anti-cancer drugs, owing to their radioprotection of normal tissue and the ability to afford pain management in cancer patients via prevention of chronic morphine tolerance.
Collapse
Affiliation(s)
- Zahid N. Rabbani
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - XiuWu Zhang
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
| | - Benjamin J. Moeller
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
| | - Sinisa Haberle
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | | | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
- Corresponding authors: Ines Batinic-Haberle, Ph. D., Department of Radiation Oncology-Cancer Biology, Duke University Medical Center, Research Drive, 281b/285 MSRB I, Box 3455, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, , Zeljko Vujaskovic, MD, PhD, Duke University Medical Center, 201 MSRB, Box 3455, Durham, NC 27710, Tel: 919-681-1675, Fax: 919-684-8718,
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710
- Corresponding authors: Ines Batinic-Haberle, Ph. D., Department of Radiation Oncology-Cancer Biology, Duke University Medical Center, Research Drive, 281b/285 MSRB I, Box 3455, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, , Zeljko Vujaskovic, MD, PhD, Duke University Medical Center, 201 MSRB, Box 3455, Durham, NC 27710, Tel: 919-681-1675, Fax: 919-684-8718,
| |
Collapse
|
25
|
Determination of residual manganese in Mn porphyrin-based superoxide dismutase (SOD) and peroxynitrite reductase mimics. J Pharm Biomed Anal 2009; 50:1088-91. [PMID: 19660888 DOI: 10.1016/j.jpba.2009.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/12/2009] [Accepted: 07/01/2009] [Indexed: 11/21/2022]
Abstract
The awareness of the beneficial effects of Mn porphyrin-based superoxide dismutase (SOD) mimics and peroxynitrite scavengers on decreasing oxidative stress injuries has increased the use of these compounds as mechanistic probes and potential therapeutics. Simple Mn2+ salts, however, have SOD-like activity in their own right both in vitro and in vivo. Thus, quantification/removal of residual Mn2+ species in Mn-based therapeutics is critical to an unambiguous interpretation of biological data. Herein we report a simple, sensitive, and specific method to determine residual Mn2+ in Mn porphyrin preparations that combines a hydrometallurgical approach for separation/speciation of metal compounds with a spectrophotometric strategy for Mn determination. The method requires only common chemicals and a spectrophotometer and is based on the extraction of residual Mn2+ by bis(2-ethylhexyl)hydrogenphosphate (D2EHPA) into kerosene, re-extraction into acid, and neutralization followed by UV-vis determination of the Mn2+ levels via a Cd2+-catalyzed metallation of the H2TCPP4- porphyrin indicator. The overall procedure is simple, sensitive, specific, and amenable to adaptation. This quantification method has been routinely used by us for a large variety of water-soluble porphyrins.
Collapse
|
26
|
Kos I, Rebouças JS, DeFreitas-Silva G, Salvemini D, Vujaskovic Z, Dewhirst MW, Spasojevic I, Batinic-Haberle I. Lipophilicity of potent porphyrin-based antioxidants: comparison of ortho and meta isomers of Mn(III) N-alkylpyridylporphyrins. Free Radic Biol Med 2009; 47:72-8. [PMID: 19361553 PMCID: PMC2694496 DOI: 10.1016/j.freeradbiomed.2009.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/25/2009] [Accepted: 04/03/2009] [Indexed: 11/24/2022]
Abstract
Mn(III) N-alkylpyridylporphyrins are among the most potent known SOD mimics and catalytic peroxynitrite scavengers and modulators of redox-based cellular transcriptional activity. In addition to their intrinsic antioxidant capacity, bioavailability plays a major role in their in vivo efficacy. Although of identical antioxidant capacity, lipophilic MnTnHex-2-PyP is up to 120-fold more efficient in reducing oxidative stress injuries than hydrophilic MnTE-2-PyP. Owing to limitations of an analytical nature, porphyrin lipophilicity has been often estimated by the thin-layer chromatographic R(f) parameter, instead of the standard n-octanol/water partition coefficient, P(OW). Herein we used a new methodological approach to finally describe the MnP lipophilicity, using the conventional log P(OW) means, for a series of biologically active ortho and meta isomers of Mn(III) N-alkylpyridylporphyrins. Three new porphyrins (MnTnBu-3-PyP, MnTnHex-3-PyP, and MnTnHep-2-PyP) were synthesized to strengthen the conclusions. The log P(OW) was linearly related to R(f) and to the number of carbons in the alkyl chain (n(C)) for both isomer series, the meta isomers being 10-fold more lipophilic than the analogous ortho porphyrins. Increasing the length of the alkyl chain by one carbon atom increases the log P(OW) value approximately 1 log unit with both isomers. Dramatic approximately 4 and approximately 5 orders of magnitude increases in the lipophilicity of the ortho isomers, by extending the pyridyl alkyl chains from two (MnTE-2-PyP, log P(OW)=-6.89) to six (MnTnHex-2-PyP, log P(OW)=-2.76) and eight carbon atoms (MnTnOct-2-PyP, log P(OW)=-1.24), parallels the increased efficacy in several oxidative-stress injury models, particularly those of the central nervous system, in which transport across the blood-brain barrier is critical. Although meta isomers are only slightly less potent SOD mimics and antioxidants than their ortho analogues, their higher lipophilicity and smaller bulkiness may lead to a higher cellular uptake and overall similar effectiveness in vivo.
Collapse
Affiliation(s)
- Ivan Kos
- Department of Radiation Oncology, University Medical School, Durham NC 27710
| | - Júlio S. Rebouças
- Department of Radiation Oncology, University Medical School, Durham NC 27710
| | | | - Daniela Salvemini
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University, St. Louis, MO 63110, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, University Medical School, Durham NC 27710
| | - Mark W. Dewhirst
- Department of Radiation Oncology, University Medical School, Durham NC 27710
| | - Ivan Spasojevic
- Department of Medicine Duke, University Medical School, Durham NC 27710
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, University Medical School, Durham NC 27710
- Corresponding author: Ines Batinic-Haberle, Ph. D., Department of Radiation Oncology-Cancer Biology, Duke University Medical Center, Research Drive, 281b/285 MSRB I, Box 3455, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, e-mail:
| |
Collapse
|
27
|
Rosenthal RA, Huffman KD, Fisette LW, Damphousse CA, Callaway WB, Malfroy B, Doctrow SR. Orally available Mn porphyrins with superoxide dismutase and catalase activities. J Biol Inorg Chem 2009; 14:979-91. [PMID: 19504132 PMCID: PMC2716445 DOI: 10.1007/s00775-009-0550-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 05/18/2009] [Indexed: 12/04/2022]
Abstract
Superoxide dismutase/catalase mimetics, such as salen Mn complexes and certain metalloporphyrins, catalytically neutralize reactive oxygen and nitrogen species, which have been implicated in the pathogenesis of many serious diseases. Both classes of mimetic are protective in animal models of oxidative stress. However, only AEOL11207 and EUK-418, two uncharged Mn porphyrins, have been shown to be orally bioavailable. In this study, EUK-418 and several new analogs (the EUK-400 series) were synthesized and shown to exhibit superoxide dismutase, catalase, and peroxidase activities in vitro. Some also protected PC12 cells against staurosporine-induced cell death. All EUK-400 compounds were stable in simulated gastric fluid, and most were substantially more lipophilic than the salen Mn complexes EUK-189 and EUK-207, which lack oral activity. Pharmacokinetics studies demonstrate the presence of all EUK-400 series compounds in the plasma of rats after oral administration. These EUK-400 series compounds are potential oral therapeutic agents for cellular damage caused by oxidative stress.
Collapse
|
28
|
Ferrer-Sueta G, Radi R. Chemical biology of peroxynitrite: kinetics, diffusion, and radicals. ACS Chem Biol 2009; 4:161-77. [PMID: 19267456 DOI: 10.1021/cb800279q] [Citation(s) in RCA: 537] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Peroxynitrite is formed by the very fast reaction of nitric oxide and superoxide radicals, a reaction that kinetically competes with other routes that chemically consume or physically sequester the reagents. It can behave either as an endogenous cytotoxin toward host tissues or a cytotoxic effector molecule against invading pathogens, depending on the cellular source and pathophysiological setting. Peroxynitrite is in itself very reactive against a few specific targets that range from efficient detoxification systems, such as peroxiredoxins, to reactions eventually leading to enhanced radical formation (e.g., nitrogen dioxide and carbonate radicals), such as the reaction with carbon dioxide. Thus, the chemical biology of peroxynitrite is dictated by the chemical kinetics of its formation and decay and by the diffusion across membranes of the species involved, including peroxynitrite itself. On the other hand, most durable traces of peroxynitrite passing (such as 3-nitrotyrosine) are derived from radicals formed from peroxynitrite by routes that represent extremely low-yield processes but that have potentially critical biological consequences. Here we have reviewed the chemical kinetics of peroxynitrite as a biochemical transient species in order to estimate its rates of formation and decay and then its steady-state concentration in different intra- or extracellular compartments, trying to provide a quantitative basis for its reactivity; additionally, we have considered diffusion across membranes to locate its possible effects. Finally, we have assessed the most successful attempts to intercept peroxynitrite by pharmacological intervention in their potential to increment the existing biological defenses that routinely deal with this cytotoxin.
Collapse
Affiliation(s)
- Gerardo Ferrer-Sueta
- Laboratorio de Físicoquímica Biológica, Facultad de Ciencias
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
29
|
Batinić-Haberle I, Ndengele MM, Cuzzocrea S, Rebouças JS, Spasojević I, Salvemini D. Lipophilicity is a critical parameter that dominates the efficacy of metalloporphyrins in blocking the development of morphine antinociceptive tolerance through peroxynitrite-mediated pathways. Free Radic Biol Med 2009; 46:212-9. [PMID: 18983908 PMCID: PMC2656771 DOI: 10.1016/j.freeradbiomed.2008.09.037] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 09/16/2008] [Accepted: 09/30/2008] [Indexed: 12/31/2022]
Abstract
Severe pain syndromes reduce the quality of life of patients with inflammatory and neoplastic diseases, partly because reduced analgesic effectiveness with chronic opiate therapy (i.e., tolerance) leads to escalating doses and distressing side effects. Peroxynitrite-mediated nitroxidative stress in the dorsal horn of the spinal cord plays a critical role in the induction and development of antinociceptive tolerance to morphine. This provides a valid pharmacological basis for developing peroxynitrite scavengers as potent adjuncts to opiates in the management of pain. The cationic Mn(III) ortho-N-alkylpyridylporphyrins MnTE-2-PyP(5+) and MnTnHex-2-PyP(5+) are among the most potent peroxynitrite scavengers, with nearly identical scavenging rate constants (approximately 10(7) M(-1) s(-1)). Yet, MnTnHex-2-PyP(5+) is significantly more lipophilic and more bioavailable and, in turn, was 30-fold more effective in blocking the development of morphine antinociceptive tolerance than MnTE-2-PyP(5+) using the hot-plate test in a well-characterized murine model. The hydrophilic MnTE-2-PyP(5+) and the lipophilic MnTnHex-2-PyP(5+) were 10- and 300-fold, respectively, more effective in inhibiting morphine tolerance than the hydrophilic Fe(III) porphyrin FeTM-4-PyP(5+). Both Mn porphyrins decreased levels of TNF-alpha, IL-1 beta, and IL-6 to normal values. Neither of them affected acute morphine antinociceptive effects nor caused motor function impairment. Also neither was able to reverse already established morphine tolerance. We have recently shown that the anionic porphyrin Mn(III) tetrakis(4-carboxylatophenyl)porphyrin is selective in removing ONOO(-) over O(2)(-), but at approximately 2 orders of magnitude lower efficacy than MnTE-2-PyP(5+) and MnTnHex-2-PyP(5+), which in turn parallels up to 100-fold lower ability to reverse morphine tolerance. These data (1) support the role of peroxynitrite rather than superoxide as a major mechanism in blocking the development of morphine tolerance and (2) show that lipophilicity is a critical parameter in enhancing the potency of such novel peroxynitrite scavengers.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA
- Address correspondence to: Daniela Salvemini, PhD, Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University School of Medicine, St Louis, USA Phone: (1) 314. 577. 8856; Fax (1) 314. 577. 8859 e-mail: and Ines Batinić-Haberle, PhD, Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA, tel: (1) 919. 684. 2101, Fax: (1) 919. 684. 8718, e-mail:
| | - Michael M. Ndengele
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, 1402 S. Grand Blvd., Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | - Salvatore Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Italy
- IRCCS Centro Neurolesi “Bonino-Pulejo” Messina, Italy
| | - Júlio. S. Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA
| | - Ivan Spasojević
- Department of Medicine, Duke University Medical School, Durham NC 27710, USA
| | - Daniela Salvemini
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, 1402 S. Grand Blvd., Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Address correspondence to: Daniela Salvemini, PhD, Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University School of Medicine, St Louis, USA Phone: (1) 314. 577. 8856; Fax (1) 314. 577. 8859 e-mail: and Ines Batinić-Haberle, PhD, Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA, tel: (1) 919. 684. 2101, Fax: (1) 919. 684. 8718, e-mail:
| |
Collapse
|
30
|
Batinić-Haberle I, Cuzzocrea S, Rebouças JS, Ferrer-Sueta G, Mazzon E, Di Paola R, Radi R, Spasojević I, Benov L, Salvemini D. Pure MnTBAP selectively scavenges peroxynitrite over superoxide: comparison of pure and commercial MnTBAP samples to MnTE-2-PyP in two models of oxidative stress injury, an SOD-specific Escherichia coli model and carrageenan-induced pleurisy. Free Radic Biol Med 2009; 46:192-201. [PMID: 19007878 PMCID: PMC2742324 DOI: 10.1016/j.freeradbiomed.2008.09.042] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 09/14/2008] [Accepted: 09/30/2008] [Indexed: 12/20/2022]
Abstract
MnTBAP is often referred to as an SOD mimic in numerous models of oxidative stress. We have recently reported that pure MnTBAP does not dismute superoxide, but commercial or poorly purified samples are able to perform O2.- dismutation with low-to-moderate efficacy via non-innocent Mn-containing impurities. Herein, we show that neither commercial nor pure MnTBAP could substitute for SOD enzyme in a SOD-deficient Escherichia coli model, whereas MnTE-2-PyP-treated SOD-deficient E. coli grew as well as a wild-type strain. This SOD-specific system indicates that MnTBAP does not act as an SOD mimic in vivo. In another model, carrageenan-induced pleurisy in mice, inflammation was evidenced by increased pleural fluid exudate and neutrophil infiltration and activation: these events were blocked by 0.3 mg/kg MnTE-2-PyP and, to a slightly lesser extent, by 10 mg/kg of either MnTBAP. Also, 3-nitrotyrosine formation, an indication of peroxynitrite existence in vivo, was blocked by both compounds; again MnTE-2-PyP was 33-fold more effective. Pleurisy model data indicate that MnTBAP exerts some protective actions in common with MnTE-2-PyP, which are not O2.- related and can be fully rationalized if one considers that the common biological role shared by MnTBAP and MnTE-2-PyP is related to their reduction of peroxynitrite and carbonate radical, the latter arising from ONOOCO2 adduct. The log kcat (O2.-) value for MnTBAP is estimated to be about 3.16, which is approximately 5 and approximately 6 orders of magnitude smaller than the SOD activities of the potent SOD mimic MnTE-2-PyP and Cu,Zn-SOD, respectively. This very low value indicates that MnTBAP is too inefficient at dismuting superoxide to be of any biological impact, which was confirmed in the SOD-deficient E. coli model. The peroxynitrite scavenging ability of MnTBAP, however, is only approximately 2.5 orders of magnitude smaller than that of MnTE-2-PyP and is not significantly affected by the presence of the SOD-active impurities in the commercial MnTBAP sample (log k red (ONOO-) = 5.06 for pure and 4.97 for commercial sample). The reduction of carbonate radical is equally fast with MnTBAP and MnTE-2-PyP. The dose of MnTBAP required to yield oxidative stress protection and block nitrotyrosine formation in the pleurisy model is > 1.5 orders of magnitude higher than that of MnTE-2-PyP, which could be related to the lower ability of MnTBAP to scavenge peroxynitrite. The slightly better protection observed with the commercial MnTBAP sample (relative to the pure MnTBAP) could arise from its impurities, which, by scavenging O2.-, reduce consequently the overall peroxynitrite and secondary ROS/RNS levels. These observations have profound biological repercussions as they may suggest that the effect of MnTBAP observed in numerous studies may conceivably relate to peroxynitrite scavenging. Moreover, provided that pure MnTBAP is unable to dismute superoxide at any significant extent, but is able to partially scavenge peroxynitrite and carbonate radical, this compound may prove valuable in distinguishing ONOO-/CO3.- from O2.- pathways.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph.D., Duke University Medical School, Department of Radiation Oncology - Cancer Biology, 281b/285 MSRB I, Box 3455, Durham, NC 27710, Tel: +1-919-684-2101; Fax: +1-919-684-8718, e-mail: , Daniela Salvemini, Ph.D., Saint Louis University, Department of Internal Medicine - Division on Pulmonary, Critical Care and Sleep Medicine, 3635 Vista Avenue, Desloge Towers 7th Floor, St Louis, MO 6311, Tel: +1-314-577-8856; Fax: +1-314-577-8859, e-mail:
| | - Salvatore Cuzzocrea
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy and Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - Júlio S. Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham NC 27710, USA
| | - Gerardo Ferrer-Sueta
- Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias, and Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo 11400, Uruguay
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy and Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - Rosanna Di Paola
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy and Department of Clinical and Experimental Medicine and Pharmacology, School of Medicine, University of Messina, Messina, Italy
| | - Rafael Radi
- Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias, and Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo 11400, Uruguay
| | - Ivan Spasojević
- Department of Medicine, Duke University Medical School, Durham NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Safat 13110, Kuwait
| | - Daniela Salvemini
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Saint Louis University, St. Louis, MO 63110, USA
- Corresponding authors: Ines Batinic-Haberle, Ph.D., Duke University Medical School, Department of Radiation Oncology - Cancer Biology, 281b/285 MSRB I, Box 3455, Durham, NC 27710, Tel: +1-919-684-2101; Fax: +1-919-684-8718, e-mail: , Daniela Salvemini, Ph.D., Saint Louis University, Department of Internal Medicine - Division on Pulmonary, Critical Care and Sleep Medicine, 3635 Vista Avenue, Desloge Towers 7th Floor, St Louis, MO 6311, Tel: +1-314-577-8856; Fax: +1-314-577-8859, e-mail:
| |
Collapse
|
31
|
Spasojević I, Chen Y, Noel TJ, Fan P, Zhang L, Rebouças JS, St. Clair DK, Batinić-Haberle I. Pharmacokinetics of the potent redox-modulating manganese porphyrin, MnTE-2-PyP(5+), in plasma and major organs of B6C3F1 mice. Free Radic Biol Med 2008; 45:943-9. [PMID: 18598757 PMCID: PMC2583406 DOI: 10.1016/j.freeradbiomed.2008.05.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 05/04/2008] [Accepted: 05/19/2008] [Indexed: 11/16/2022]
Abstract
Mn(III) tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+), a potent catalytic superoxide and peroxynitrite scavenger, has been beneficial in several oxidative stress-related diseases thus far examined. Pharmacokinetic studies are essential for the better assessment of the therapeutic potential of MnTE-2-PyP(5+) and similar compounds, as well as for the modulation of their bioavailability and toxicity. Despite high hydrophilicity, this drug entered mitochondria after a single 10 mg/kg intraperitoneal injection at levels high enough (5.1 muM; 2.95 ng/mg protein) to protect against superoxide/peroxynitrite damage. Utilizing the same analytical approach, which involves the reduction of MnTE-2-PyP(5+) followed by the exchange of Mn(2+) with Zn(2+) and HPLC/fluorescence detection of ZnTE-2-PyP(4+), we measured levels of MnTE-2-PyP(5+) in mouse plasma, liver, kidney, lung, heart, spleen, and brain over a period of 7 days after a single intraperitoneal injection of 10 mg/kg. Two B6C3F1 female mice per time point were used. The pharmacokinetic profile in plasma and organs was complex; thus a noncompartmental approach was utilized to calculate the area under the curve, c(max), t(max), and drug elimination half-time (t(1/2)). In terms of levels of MnTE-2-PyP(5+) found, the organs can be classified into three distinct groups: (1) high levels (kidney, liver, and spleen), (2) moderate levels (lung and heart), and (3) low levels (brain). The maximal levels in plasma, kidney, spleen, lung, and heart are reached within 45 min, whereas in the case of liver a prolonged absorption phase was observed, with the maximal concentration reached at 8 h. Moreover, accumulation of the drug in brain continued beyond the time of the experiment (7 days) and is likely to be driven by the presence of negatively charged phospholipids. For tissues other than brain, a slow elimination phase (single exponential decay, t(1/2)=60 to 135 h) was observed. The calculated pharmacokinetic parameters will be used to design optimal dosing regimens in future preclinical studies utilizing this and similar compounds.
Collapse
Affiliation(s)
- Ivan Spasojević
- Department of Medicine, Duke University Medical School, Durham, NC 27710
- Corresponding authors: Ivan Spasojević Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 684-8311, Fax: 684-9094, e-mail: , Ines Batinić-Haberle, Department of Radiation Oncology, Duke University Medical Center, 231 Nanaline H. Duke, Box 3711, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8885, e-mail:
| | - Yumin Chen
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY, 40536
| | - Teresa J. Noel
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY, 40536
| | - Ping Fan
- Department of Medicine, Duke University Medical School, Durham, NC 27710
| | - Lichun Zhang
- Department of Medicine, Duke University Medical School, Durham, NC 27710
| | - Julio S. Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710
| | - Daret K. St. Clair
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY, 40536
| | - Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710
- Corresponding authors: Ivan Spasojević Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 684-8311, Fax: 684-9094, e-mail: , Ines Batinić-Haberle, Department of Radiation Oncology, Duke University Medical Center, 231 Nanaline H. Duke, Box 3711, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8885, e-mail:
| |
Collapse
|
32
|
Silva DCD, DeFreitas-Silva G, Nascimento ED, Rebouças JS, Barbeira PJS, Carvalho MEMDD, Idemori YM. Spectral, electrochemical, and catalytic properties of a homologous series of manganese porphyrins as cytochrome P450 model: The effect of the degree of β-bromination. J Inorg Biochem 2008; 102:1932-41. [DOI: 10.1016/j.jinorgbio.2008.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 11/30/2022]
|
33
|
DeFreitas-Silva G, Rebouças JS, Spasojevi I, Benov L, Idemori YM, -Haberle IB. SOD-like activity of Mn(II) beta-octabromo-meso-tetrakis(N-methylpyridinium-3-yl)porphyrin equals that of the enzyme itself. Arch Biochem Biophys 2008; 477:105-12. [PMID: 18477465 PMCID: PMC2577908 DOI: 10.1016/j.abb.2008.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2008] [Revised: 04/21/2008] [Accepted: 04/24/2008] [Indexed: 11/29/2022]
Abstract
Mn porphyrins are among the most efficient SOD mimics with potency approaching that of SOD enzymes. The most potent ones, Mn(III) N-alkylpyridylporphyrins bear positive charges in a close proximity to the metal site, affording thermodynamic and kinetic facilitation for the reaction with negatively charged superoxide. The addition of electron-withdrawing bromines onto beta-pyrrolic positions dramatically improves thermodynamic facilitation for the O2*- dismutation. We have previously characterized the para isomer, Mn(II)Br(8)TM-4-PyP(4+) [Mn(II) beta-octabromo-meso-tetrakis(N-methylpyridinium-4-yl)porphyrin]. Herein we fully characterized its meta analogue, Mn(II)Br(8)TM-3-PyP(4+) with respect to UV/vis spectroscopy, electron spray mass spectrometry, electrochemistry, O2*- dismutation, metal-ligand stability, and the ability to protect SOD-deficient Escherichia coli in comparison with its para analogue. The increased electron-deficiency of the metal center stabilizes Mn in its +2 oxidation state. The metal-centered Mn(III)/Mn(II) reduction potential, E((1/2))=+468 mV vs NHE, is increased by 416 mV with respect to non-brominated analogue, Mn(III)TM-3-PyP(5+) and is only 12 mV less positive than for para isomer. Yet, the complex is significantly more stable towards the loss of metal than its para analogue. As expected, based on the structure-activity relationships, an increase in E((1/2)) results in a higher catalytic rate constant for the O2*- dismutation, log k(cat)> or =8.85; 1.5-fold increase with respect to the para isomer. The IC(50) was calculated to be < or =3.7 nM. Manipulation of the electron-deficiency of a cationic porphyrin resulted, therefore, in the highest k(cat) ever reported for a metalloporphyrin, being essentially identical to the k(cat) of superoxide dismutases (log k(cat)=8.84-9.30). The positive kinetic salt effect points to the unexpected, unique and first time recorded behavior of Mn beta-octabrominated porphyrins when compared to other Mn porphyrins studied thus far. When species of opposing charges react, the increase in ionic strength invariably results in the decreased rate constant; with brominated porphyrins the opposite was found to be true. The effect is 3.5-fold greater with meta than with para isomer, which is discussed with respect to the closer proximity of the quaternary nitrogens of the meta isomer to the metal center than that of the para isomer. The potency of Mn(II)Br(8)TM-3-PyP(4+) was corroborated by in vivo studies, where 500 nM allows SOD-deficient E. coli to grow >60% of the growth of wild type; at concentrations > or =5 microM it exhibits toxicity. Our work shows that exceptionally high k(cat) for the O2*- disproportionation can be achieved not only with an N(5)-type coordination motif, as rationalized previously for aza crown ether (cyclic polyamines) complexes, but also with a N(4)-type motif as in the Mn porphyrin case; both motifs sharing "up-down-up-down" steric arrangement.
Collapse
Affiliation(s)
- Gilson DeFreitas-Silva
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Júlio S. Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710, USA
| | - Ivan Spasojevi
- Department of Medicine, Duke University Medical School, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Safat, 13110, Kuwait
| | - Ynara M. Idemori
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ines Batini -Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710, USA
| |
Collapse
|
34
|
Rebouças JS, Spasojević I, Batinić-Haberle I. Quality of potent Mn porphyrin-based SOD mimics and peroxynitrite scavengers for pre-clinical mechanistic/therapeutic purposes. J Pharm Biomed Anal 2008; 48:1046-9. [PMID: 18804338 DOI: 10.1016/j.jpba.2008.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Revised: 07/28/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
Abstract
Cationic Mn porphyrins are among the most potent SOD mimics and peroxynitrite scavengers. They have been widely and successfully used in different models of oxidative stress and are either progressing towards or are in phase I of clinical trials. The most frequently used compounds are Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+) or AEOL10113), its methyl analogue (MnTM-2-PyP(5+) or AEOL10112), and Mn(III) meso-tetrakis(4-benzoic acid)porphyrin (MnTBAP). A great discrepancy between the in vivo data obtained with Calbiochem preparations and those of authentic MnTE-2-PyP(5+) and MnTM-2-PyP(5+) samples were recently observed. Surprisingly, the commercial samples were invariably of poor identity and consisted of mixtures of nearly equal contributions of non-alkylated, mono-, di-, tri- and tetraalkylated porphyrins, lacking thus the major structural entity that determines their antioxidant potency, i.e., the four positively charged orthoN-alkylpyridyl groups that afford thermodynamic tuning of the active site and electrostatic guidance of anionic superoxide and peroxynitrite species toward the metal center. The MnTE-2-PyP(5+) and MnTM-2-PyP(5+) compounds were not even the major species in the commercial samples sold as "MnTE-2-PyP" and "MnTM-2-PyP", respectively. While we have already reported the insufficient impurity of the MnTBAP samples from Alexis and other suppliers, in one more recent lot the situation is dramatic, as 25% of the sample was not MnTBAP, but metal-free ligand, H(2)TBAP. The (unintentional) use of the Mn porphyrins of low quality compromises therapeutic and/or mechanistic conclusions. Simple techniques, which include thin-layer chromatography, electrospray-mass spectrometry, UV-vis spectroscopy, and electrochemistry described here could be used routinely to check the overall quality of Mn porphyrins in order to avoid misleading conclusions and waste of valuable resources (animals, compounds, time, manpower).
Collapse
Affiliation(s)
- Júlio S Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710, USA
| | | | | |
Collapse
|
35
|
Rebouças JS, DeFreitas-Silva G, Spasojević I, Idemori YM, Benov L, Batinić-Haberle I. Impact of electrostatics in redox modulation of oxidative stress by Mn porphyrins: protection of SOD-deficient Escherichia coli via alternative mechanism where Mn porphyrin acts as a Mn carrier. Free Radic Biol Med 2008; 45:201-10. [PMID: 18457677 PMCID: PMC2614336 DOI: 10.1016/j.freeradbiomed.2008.04.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 04/03/2008] [Accepted: 04/05/2008] [Indexed: 01/11/2023]
Abstract
Understanding the factors that determine the ability of Mn porphyrins to scavenge reactive species is essential for tuning their in vivo efficacy. We present herein the revised structure-activity relationships accounting for the critical importance of electrostatics in the Mn porphyrin-based redox modulation systems and show that the design of effective SOD mimics (per se) based on anionic porphyrins is greatly hindered by inappropriate electrostatics. A new strategy for the beta-octabromination of the prototypical anionic Mn porphyrins Mn(III) meso-tetrakis(p-carboxylatophenyl)porphyrin ([Mn(III)TCPP](3-) or MnTBAP(3-)) and Mn(III) meso-tetrakis(p-sulfonatophenyl)porphyrin ([Mn(III)TSPP](3-)), to yield the corresponding anionic analogues [Mn(III)Br(8)TCPP](3-) and [Mn(III)Br(8)TSPP](3-), respectively, is described along with characterization data, stability studies, and their ability to substitute for SOD in SOD-deficient Escherichia coli. Despite the Mn(III)/Mn(II) reduction potential of [Mn(III)Br(8)TCPP](3-) and [Mn(III)Br(8)TSPP](3-) being close to the SOD-enzyme optimum and nearly identical to that of the cationic Mn(III) meso-tetrakis(N-methylpyridinium-2-yl)porphyrin (Mn(III)TM-2-PyP(5+)), the SOD activity of both anionic brominated porphyrins ([Mn(III)Br(8)TCPP](3-), E(1/2)=+213 mV vs NHE, log k(cat)=5.07; [Mn(III)Br(8)TSPP](3-), E(1/2)=+209 mV, log k(cat)=5.56) is considerably lower than that of Mn(III)TM-2-PyP(5+) (E(1/2)=+220 mV, log k(cat)=7.79). This illustrates the impact of electrostatic guidance of O(2)(-) toward the metal center of the mimic. With low k(cat), the [Mn(III)TCPP](3-), [Mn(III)TSPP](3-), and [Mn(III)Br(8)TCPP](3-) did not rescue SOD-deficient E. coli. The striking ability of [Mn(III)Br(8)TSPP](3-) to substitute for the SOD enzymes in the E. coli model does not correlate with its log k(cat). In fact, the protectiveness of [Mn(III)Br(8)TSPP](3-) is comparable to or better than that of the potent SOD mimic Mn(III)TM-2-PyP(5+), even though the dismutation rate constant of the anionic complex is 170-fold smaller. Analyses of the medium and E. coli cell extract revealed that the major species in the [Mn(III)Br(8)TSPP](3-) system is not the Mn complex, but the free-base porphyrin [H(2)Br(8)TSPP](4-) instead. Control experiments with extracellular MnCl(2) showed the lack of E. coli protection, indicating that "free" Mn(2+) cannot enter the cell to a significant extent. We proposed herein the alternative mechanism where a labile Mn porphyrin [Mn(III)Br(8)TSPP](3-) is not an SOD mimic per se but carries Mn into the E. coli cell.
Collapse
Affiliation(s)
- Júlio S. Rebouças
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710, USA
| | - Gilson DeFreitas-Silva
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ivan Spasojević
- Department of Medicine, Duke University Medical School, Durham, NC 27710, USA
| | - Ynara M. Idemori
- Departamento de Química, ICEx, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Safat, 13110, Kuwait
| | - Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, NC 27710, USA
| |
Collapse
|
36
|
Facile synthesis of ortho-pyridyl-substituted corroles and molecular structures of analogous porphyrins. Tetrahedron Lett 2008. [DOI: 10.1016/j.tetlet.2008.04.113] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Ivanović-Burmazović I, van Eldik R. Metal complex-assisted activation of small molecules. From NO to superoxide and peroxides. Dalton Trans 2008:5259-75. [DOI: 10.1039/b805450a] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|