1
|
Soares RB, Pinto J, Amaro F, Manguinhas R, Gil N, Rosell R, Batinic-Haberle I, Fernandes AS, Oliveira NG, Guedes de Pinho P. Impact of the redox-active MnTnHex-2-PyP 5+ and cisplatin on the metabolome of non-small cell lung cancer cells. Biochem Pharmacol 2024; 227:116424. [PMID: 39004232 DOI: 10.1016/j.bcp.2024.116424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/07/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Redox-based cancer therapeutic strategies aim to raise reactive oxygen species (ROS) levels in cancer cells, thus modifying their redox status, and eventually inducing cell death. Promising compounds, known as superoxide dismutase mimics (SODm), e.g. MnTnHex-2-Py5+ (MnTnHex), could increase intracellular H2O2 in cancer cells with deficient ROS removal systems and therefore enhance radio- and chemotherapy efficacy. We have previously shown that MnTnHex was cytotoxic either alone or combined with cisplatin to non-small cell lung cancer (NSCLC) cells. To gain a deeper understanding of the effects and safety of this compound, it is crucial to analyze the metabolic alterations that take place within the cell. Our goal was thus to study the intracellular metabolome (intracellular metabolites) of NSCLC cells (A549 and H1975) using nuclear magnetic resonance (NMR) spectroscopy-based metabolomics to evaluate the changes in cellular metabolism upon exposure to MnTnHex per se or in combination with cisplatin. 1H NMR metabolomics revealed a higher number of significantly altered metabolites in A549 cells exposed to MnTnHex alone or combined with cisplatin in comparison with non-treated cells (nine dysregulated metabolites), suggesting an impact on aminoacyl-tRNA biosynthesis, glycolysis/gluconeogenesis, taurine, hypotaurine, glycerophospholipid, pyruvate, arginine and proline metabolisms. Regarding H1975 cells, significant alterations in the levels of six metabolites were observed upon co-treatment with MnTnHex and cisplatin, suggesting dysregulations in aminoacyl-tRNA biosynthesis, arginine and proline metabolism, pyruvate metabolism, and glycolysis/gluconeogenesis. These findings help us to understand the impact of MnTnHex on NSCLC cells. Importantly, specific altered metabolites, such as taurine, may contribute to the chemosensitizing effects of MnTnHex.
Collapse
Affiliation(s)
- Rita B Soares
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal; Lung Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Lab. of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Filipa Amaro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Lab. of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Manguinhas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Nuno Gil
- Lung Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Rafael Rosell
- Dr. Rosell Oncology Institute, 08028 Barcelona, Spain; Institute Germans Trias i Pujol, 08916 Badalona, Barcelona, Spain
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ana S Fernandes
- Universidade Lusófona's Research Center for Biosciences & Health Technologies (CBIOS), Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Lab. of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
Dos Anjos Cordeiro JM, Santos LC, Santos BR, de Jesus Nascimento AE, Santos EO, Barbosa EM, de Macêdo IO, Mendonça LD, Sarmento-Neto JF, Pinho CS, Coura ETDS, Santos ADS, Rodrigues ME, Rebouças JS, De-Freitas-Silva G, Munhoz AD, de Lavor MSL, Silva JF. Manganese porphyrin-based treatment improves fetal-placental development and protects against oxidative damage and NLRP3 inflammasome activation in a rat maternal hypothyroidism model. Redox Biol 2024; 74:103238. [PMID: 38870780 PMCID: PMC11225907 DOI: 10.1016/j.redox.2024.103238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/15/2024] Open
Abstract
Oxidative stress (OS) and endoplasmic reticulum stress (ERS) are at the genesis of placental disorders observed in preeclampsia, intrauterine growth restriction, and maternal hypothyroidism. In this regard, cationic manganese porphyrins (MnPs) comprise potent redox-active therapeutics of high antioxidant and anti-inflammatory potential, which have not been evaluated in metabolic gestational diseases yet. This study evaluated the therapeutic potential of two MnPs, [MnTE-2-PyP]5+ (MnP I) and [MnT(5-Br-3-E-Py)P]5+ (MnP II), in the fetal-placental dysfunction of hypothyroid rats. Hypothyroidism was induced by administration of 6-Propyl-2-thiouracil (PTU) and treatment with MnPs I and II 0.1 mg/kg/day started on the 8th day of gestation (DG). The fetal and placental development, and protein and/or mRNA expression of antioxidant mediators (SOD1, CAT, GPx1), hypoxia (HIF1α), oxidative damage (8-OHdG, MDA), ERS (GRP78 and CHOP), immunological (TNFα, IL-6, IL-10, IL-1β, IL-18, NLRP3, Caspase1, Gasdermin D) and angiogenic (VEGF) were evaluated in the placenta and decidua on the 18th DG using immunohistochemistry and qPCR. ROS and peroxynitrite (PRX) were quantified by fluorometric assay, while enzyme activities of SOD, GST, and catalase were evaluated by colorimetric assay. MnPs I and II increased fetal body mass in hypothyroid rats, and MnP I increased fetal organ mass. MnPs restored the junctional zone morphology in hypothyroid rats and increased placental vascularization. MnPs blocked the increase of OS and ERS mediators caused by hypothyroidism, showing similar levels of expression of HIFα, 8-OHdG, MDA, Gpx1, GRP78, and Chop to the control. Moreover, MnPs I and/or II increased the protein expression of SOD1, Cat, and GPx1 and restored the expression of IL10, Nlrp3, and Caspase1 in the decidua and/or placenta. However, MnPs did not restore the low placental enzyme activity of SOD, CAT, and GST caused by hypothyroidism, while increased the decidual and placental protein expression of TNFα. The results show that treatment with MnPs improves the fetal-placental development and the placental inflammatory state of hypothyroid rats and protects against oxidative stress and reticular stress caused by hypothyroidism at the maternal-fetal interface.
Collapse
Affiliation(s)
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | | | - Emilly Oliveira Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Erikles Macêdo Barbosa
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Isabela Oliveira de Macêdo
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Letícia Dias Mendonça
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - José Ferreira Sarmento-Neto
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, Joao Pessoa, Brazil
| | - Clarice Santos Pinho
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Acácio de Sá Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Marciel Elio Rodrigues
- Departamento de Ciências Exatas e Tecnológicas, Universidade Estadual Do Sudoeste da Bahia, Vitória da Conquista, Brazil
| | - Júlio Santos Rebouças
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, Joao Pessoa, Brazil
| | - Gilson De-Freitas-Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre Dias Munhoz
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Mário Sérgio Lima de Lavor
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Brazil.
| |
Collapse
|
3
|
Soares RB, Manguinhas R, Costa JG, Saraiva N, Gil N, Rosell R, Camões SP, Batinic-Haberle I, Spasojevic I, Castro M, Miranda JP, Amaro F, Pinto J, Fernandes AS, Guedes de Pinho P, Oliveira NG. MnTnHex-2-PyP 5+ Displays Anticancer Properties and Enhances Cisplatin Effects in Non-Small Cell Lung Cancer Cells. Antioxidants (Basel) 2022; 11:2198. [PMID: 36358570 PMCID: PMC9686800 DOI: 10.3390/antiox11112198] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 08/13/2023] Open
Abstract
The manganese(III) porphyrin MnTnHex-2-PyP5+ (MnTnHex) is a potent superoxide dismutase mimic and modulator of redox-based transcriptional activity that has been studied in the context of different human disease models, including cancer. Nevertheless, for lung cancer, hardly any information is available. Thus, the present work aims to fill this gap and reports the effects of MnTnHex in non-small cell lung cancer (NSCLC) cells, more specifically, A549 and H1975 cells, in vitro. Both cell lines were initially characterized in terms of innate levels of catalase, glutathione peroxidase 1, and peroxiredoxins 1 and 2. To assess the effect of MnTnHex in NSCLC, alone or in combination with cisplatin, endpoints related to the cell viability, cell cycle distribution, cell motility, and characterization of the volatile carbonyl compounds (VCCs) generated in the extracellular medium (i.e., exometabolome) were addressed. The results show that MnTnHex as a single drug markedly reduced the viability of both NSCLC cell lines, with some IC50 values reaching sub-micromolar levels. This redox-active drug also altered the cell cycle distribution, induced cell death, and increased the cytotoxicity pattern of cisplatin. MnTnHex also reduced collective cell migration. Finally, the metabolomics study revealed an increase in the levels of a few VCCs associated with oxidative stress in MnTnHex-treated cells. Altogether these results suggest the therapeutic potential of MnTnHex to be further explored, either alone or in combination therapy with cisplatin, in NSCLC.
Collapse
Affiliation(s)
- Rita B. Soares
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rita Manguinhas
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - João G. Costa
- Universidade Lusófona’s Research Center for Biosciences & Health Technologies (CBIOS), Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Nuno Saraiva
- Universidade Lusófona’s Research Center for Biosciences & Health Technologies (CBIOS), Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Nuno Gil
- Lung Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Rafael Rosell
- Laboratory of Cellular and Molecular Biology, Institute for Health Science Research Germans Trias i Pujol (IGTP), Campus Can Ruti, Ctra de Can Ruti, Camí de les Escoles, s/n, 08916 Badalona, Barcelona, Spain
| | - Sérgio P. Camões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine and PK/PD Core Laboratory, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Filipa Amaro
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana S. Fernandes
- Universidade Lusófona’s Research Center for Biosciences & Health Technologies (CBIOS), Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Biological Sciences, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
4
|
Victória HFV, Ferreira DC, Filho JBG, Martins DCS, Pinheiro MVB, Sáfar GDAM, Krambrock K. Detection of singlet oxygen by EPR: The instability of the nitroxyl radicals. Free Radic Biol Med 2022; 180:143-152. [PMID: 34979255 DOI: 10.1016/j.freeradbiomed.2021.12.303] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022]
Abstract
The use of spin traps and redox probes coupled with electron paramagnetic resonance (EPR) is a method frequently applied in the evaluation of the efficiency of photosensitizers and photocatalysts in phototherapeutic and photocatalytic processes that involve reactive oxygen species. In this way, the method helps to clarify the mechanism behind photo-induced reactions. Hydroxy-TEMP is a very specific redox probe for selectively identifying and quantifying singlet oxygen (1O2). In this work, the kinetics of radical generated by the oxidation products of the Hydroxy-TEMP redox probe was analyzed from EPR spectra in aqueous solutions of several water-soluble porphyrins ([H2T4MPyP](OTs)4, Na4[H2T4SPP], [H2T2MPyP](OTs)4, [ZnT4MyPyP](OTs)4, [MnT4MyPyP](OTs)5, H2T4CPP, and [H2T4TriMAPP](OTs)4) under white light illumination. Different factors such as the concentration of the redox probe, pH of the medium, and photostability of the porphyrins were evaluated. A systematic study was carried out to reveal the factors associated with stable radical degradation (TEMPOL) by illumination in the visible spectral region in systems containing photosensitizer (porphyrin) and redox probe (Hydroxy-TEMP). With the aid of EPR and gas chromatography coupled with mass spectroscopy (GC-MS) techniques, the mechanism of the radical degradation and the photobleaching of porphyrins were investigated. After successive interactions with the porphyrin in its excited state, in alkaline aqueous solution (pH > 10), the free radical TEMPOL is transformed into TEMPONE until the final diamagnetic product Phorone. A protocol was elaborated to identify and quantify the generation of 1O2 by Hydroxy-TEMP reliably, to avoid possible errors in the interpretation of efficiency of photosensitizers.
Collapse
Affiliation(s)
- Henrique F V Victória
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| | - Daniele C Ferreira
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - José B G Filho
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Dayse C S Martins
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Maurício V B Pinheiro
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Gustavo de A M Sáfar
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Klaus Krambrock
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
5
|
Evaluation of the compounds commonly known as superoxide dismutase and catalase mimics in cellular models. J Inorg Biochem 2021; 219:111431. [PMID: 33798828 DOI: 10.1016/j.jinorgbio.2021.111431] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022]
Abstract
Oxidative stress that results from an imbalance between the concentrations of reactive species (RS) and antioxidant defenses is associated with many pathologies. Superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase are among the key enzymes that maintain the low nanomolar physiological concentrations of superoxide and hydrogen peroxide. The increase in the levels of these species and their progeny could have deleterious effects. In this context, chemists have developed SOD and CAT mimics to supplement them when cells are overwhelmed with oxidative stress. However, the beneficial activity of such molecules in cells depends not only on their intrinsic catalytic activities but also on their stability in biological context, their cell penetration and their cellular localization. We have employed cellular assays to characterize several compounds that possess SOD and CAT activities and have been frequently used in cellular and animal models. We used cellular assays that address SOD and CAT activities of the compounds. Finally, we determined the effect of compounds on the suppression of the inflammation in HT29-MD2 cells challenged by lipopolysaccharide. When the assay requires penetration inside cells, the SOD mimics Mn(III) meso-tetrakis(N-(2'-n-butoxyethyl)pyridinium-2-yl)porphyrin (MnTnBuOE-2-PyP5+) and Mn(II) dichloro[(4aR,13aR,17aR,21aR)-1,2,3,4,4a,5,6,12,13,13a,14,15,16,17,17a,18,19,20,21,21a-eicosahydro-11,7-nitrilo-7Hdibenzo[b,h] [1,4, 7,10] tetraazacycloheptadecine-κN5,κN13,κN18,κN21,κN22] (Imisopasem manganese, M40403, CG4419) were found efficacious at 10 μM, while Mn(II) chloro N-(phenolato)-N,N'-bis[2-(N-methyl-imidazolyl)methyl]-ethane-1,2-diamine (Mn1) requires an incubation at 100 μM. This study thus demonstrates that MnTnBuOE-2-PyP5+, M40403 and Mn1 were efficacious in suppressing inflammatory response in HT29-MD2 cells and such action appears to be related to their ability to enter the cells and modulate reactive oxygen species (ROS) levels.
Collapse
|
6
|
Batinic-Haberle I, Tovmasyan A, Huang Z, Duan W, Du L, Siamakpour-Reihani S, Cao Z, Sheng H, Spasojevic I, Alvarez Secord A. H 2O 2-Driven Anticancer Activity of Mn Porphyrins and the Underlying Molecular Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6653790. [PMID: 33815656 PMCID: PMC7987459 DOI: 10.1155/2021/6653790] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Mn(III) ortho-N-alkyl- and N-alkoxyalkyl porphyrins (MnPs) were initially developed as superoxide dismutase (SOD) mimics. These compounds were later shown to react with numerous reactive species (such as ONOO-, H2O2, H2S, CO3 •-, ascorbate, and GSH). Moreover, the ability of MnPs to oxidatively modify activities of numerous proteins has emerged as their major mechanism of action both in normal and in cancer cells. Among those proteins are transcription factors (NF-κB and Nrf2), mitogen-activated protein kinases, MAPKs, antiapoptotic bcl-2, and endogenous antioxidative defenses. The lead Mn porphyrins, namely, MnTE-2-PyP5+ (BMX-010, AEOL10113), MnTnBuOE-2-PyP5+ (BMX-001), and MnTnHex-2-PyP5+, were tested in numerous injuries of normal tissue and cellular and animal cancer models. The wealth of the data led to the progression of MnTnBuOE-2-PyP5+ into four Phase II clinical trials on glioma, head and neck cancer, anal cancer, and multiple brain metastases, while MnTE-2-PyP5+ is in Phase II clinical trial on atopic dermatitis and itch.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zhiqing Huang
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Weina Duan
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Li Du
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Zhipeng Cao
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Huaxin Sheng
- Departments of Anesthesiology, Neurobiology, and Neurosurgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Pharmacokinetics/Pharmacodynamics (PK/PD) Core Laboratory, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Angeles Alvarez Secord
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
7
|
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
|
8
|
Sin KR, Ko SG, Kim CJ, Maeng TW, Choe SJ, Ri KR. Reduction of peroxynitrite by some manganoporphyrins of AEOL series: DFT approach with dispersion correction and NBO analysis. J Inorg Biochem 2020; 214:111299. [PMID: 33152662 DOI: 10.1016/j.jinorgbio.2020.111299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 11/18/2022]
Abstract
The AEOL series of manganoporphyrins (MnP; AEOL compounds were named by US Aeolus pharmaceuticals) designed as superoxide dismutase mimic are well-known for their powerful catalytic activity to neutralize reactive oxygen and nitrogen species. Reductive oxygen atom cleavage from peroxynitrite (ONOO-) to form NO2 in aqueous solution by some AEOL compounds (AEOL-10113, AEOL-10150, AEOL-11114 and AEOL-11203) was studied by DFT/M06-2X computations with D3 dispersion correction and gCP (geometrical counterpoise correction) for basis set superposition error. DFT computation showed that AEOL-10150 can form the most stable association complex {MnP…OONO} among four AEOL models. AEOL-10150 complex with ONOO- has the lowest deformation energy. In AEOL compounds and their association complexes with ONOO-, Mn atom prefered the high spin state (S = 2) to the intermediate spin state (S = 1). Natural bond orbital analysis showed that electron transfer from the most negative oxygen atom in ONOO- to Mn atom in MnP has the biggest interaction energy among all kinds of donor-acceptor interactions between ONOO- and MnP.
Collapse
Affiliation(s)
- Kye-Ryong Sin
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea.
| | - Sun-Gyong Ko
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Chol-Jin Kim
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Tae-Won Maeng
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Sung-Jub Choe
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| | - Kum-Ryong Ri
- Faculty of Chemistry, Kim Il Sung University, Pyongyang, Democratic People's Republic of Korea
| |
Collapse
|
9
|
Mourzina YG, Offenhäusser A. Electrochemical properties and biomimetic activity of water-soluble meso-substituted Mn(III) porphyrin complexes in the electrocatalytic reduction of hydrogen peroxide. J Electroanal Chem (Lausanne) 2020. [DOI: 10.1016/j.jelechem.2020.114159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
10
|
Gad SC, Sullivan DW, Mujer CV, Spainhour CB, Crapo JD. Nonclinical Safety and Toxicokinetics of MnTE-2-PyP (BMX-010), a Topical Agent in Phase 2 Trials for Psoriasis and Atopic Dermatitis. Int J Toxicol 2020; 38:291-302. [PMID: 31333066 DOI: 10.1177/1091581819852325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Since our earlier publication (Gad et al, 2013), BioMimetix has advanced BMX-010 (Manganese (III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin or MnTE020PyP; CASRN 219818-60-7) into clinical development as a topical agent for the treatment of psoriasis, atopic dermatitis, and pruritus (idiopathic nonspecific itch). A multiple dose phase I study has been completed in 64 patients without any serious adverse effects. During the course of development, the formulation was initially a gel but has been modified to a cream formulation. The nonclinical safety program has been carried onward to assess preclinical risk to patients. Additional studies completed and reported here include dermal sensitization in a Guinea Pig maximization test study, 2 rabbit phototoxicity studies, a 28-day oral toxicity study in juvenile mice, a 28-day topical systemic toxicity study in Gottingen minipigs, range-finding studies, and complete embryo-fetal developmental toxicity (Segment II) studies in mice and rabbits, an ICH M7 compliant qualification of impurities using 2 (Q)SAR in silico methods, and a 14-day subcutaneous toxicity study of mice to qualify an impurity. All studies (except the (Q)SAR evaluations) were performed in accordance with Good Laboratory Practices (GLP) using Good Manufacturing Practices (GMP) drug substance. The systemic toxicity studies, with the exception of the juvenile toxicity study, included toxicokinetic evaluations, which are reported here. The phase I clinical study had 67 patient participants who received topically applied BMX-010, and there were no notable safety findings and included pharmacokinetic determinations on these patients which are also reported here. Chronic GLP toxicity studies have been initiated in the mouse (6-month oral) and minipig (9-month dermal). To date, the only observed nonclinical toxicity remains a reversible hypertension seen in mice in response to Cmax levels with a no effect threshold, and there have been no drug-related adverse effects.
Collapse
|
11
|
Batinic-Haberle I, Spasojevic I. 25 years of development of Mn porphyrins — from mimics of superoxide dismutase enzymes to thiol signaling to clinical trials: The story of our life in the USA. J PORPHYR PHTHALOCYA 2020. [DOI: 10.1142/s1088424619300283] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We have developed Mn porphyrins (MnPs) initially as mimics of superoxide dismutase (SOD) enzymes based on structure–activity relationships. Several cationic Mn porphyrins, being substituted with cationic ortho [Formula: see text]-alkyl- or alkoxyalkylpyridyl groups in meso positions of the porphyrin ring, have been identified as potential therapeutics based on their high SOD-like activity and high bioavailability. Two of those [Mn(III) meso-tetrakis([Formula: see text]-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP[Formula: see text] (BMX-010, AEOL10113) and Mn(III) meso-tetrakis(Nn-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP[Formula: see text] (BMX-001)] are now in five Phase II clinical trials. Studies of ours, and those of others, contributed to the understanding of the diverse activities of these compounds. With biologically compatible potentials and four biologically accessible oxidation states, Mn porphyrins interact with numerous reactive species, both as oxidants and reductants. Among those reactions, their abilities to (catalytically) oxidize [Formula: see text]-glutathionylate protein thiols may perhaps be their major in vivo mode of action. Via [Formula: see text]-glutathionylation, MnPs modulate actions of signaling proteins and, in turn, cellular apoptotic and proliferative pathways. During the major part of our stay in the USA, our lives have been dedicated to Mn porphyrins. Our families and especially our son and his three babies have been our inspiration not to give up on a life often burdened with hardship. It is thus our immense pleasure to see our compounds in clinical trials. Above all, we hope that our story will inspire future researchers to persevere — women in particular.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Departments of Radiation Oncology and Pharmaceutical Research Shared Resource, Duke School of Medicine, Durham NC 27710, USA
| | - Ivan Spasojevic
- Departments of Medicine and Pharmaceutical Research Shared Resource, Duke School of Medicine, Durham NC 27710, USA
- PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke School of Medicine, Durham NC 27710, USA
| |
Collapse
|
12
|
Manganese Porphyrin-Based SOD Mimetics Produce Polysulfides from Hydrogen Sulfide. Antioxidants (Basel) 2019; 8:antiox8120639. [PMID: 31842297 PMCID: PMC6943712 DOI: 10.3390/antiox8120639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Manganese-centered porphyrins (MnPs), MnTE-2-PyP5+ (MnTE), MnTnHex-2-PyP5+ (MnTnHex), and MnTnBuOE-2-PyP5+ (MnTnBuOE) have received considerable attention because of their ability to serve as superoxide dismutase (SOD) mimetics thereby producing hydrogen peroxide (H2O2), and oxidants of ascorbate and simple aminothiols or protein thiols. MnTE-2-PyP5+ and MnTnBuOE-2-PyP5+ are now in five Phase II clinical trials warranting further exploration of their rich redox-based biology. Previously, we reported that SOD is also a sulfide oxidase catalyzing the oxidation of hydrogen sulfide (H2S) to hydrogen persulfide (H2S2) and longer-chain polysulfides (H2Sn, n = 3–7). We hypothesized that MnPs may have similar actions on sulfide metabolism. H2S and polysulfides were monitored in fluorimetric assays with 7-azido-4-methylcoumarin (AzMC) and 3′,6′-di(O-thiosalicyl)fluorescein (SSP4), respectively, and specific polysulfides were further identified by mass spectrometry. MnPs concentration-dependently consumed H2S and produced H2S2 and subsequently longer-chain polysulfides. This reaction appeared to be O2-dependent. MnP absorbance spectra exhibited wavelength shifts in the Soret and Q bands characteristic of sulfide-mediated reduction of Mn. Taken together, our results suggest that MnPs can become efficacious activators of a variety of cytoprotective processes by acting as sulfide oxidation catalysts generating per/polysulfides.
Collapse
|
13
|
Costa JG, Saraiva N, Batinic-Haberle I, Castro M, Oliveira NG, Fernandes AS. The SOD Mimic MnTnHex-2-PyP 5+ Reduces the Viability and Migration of 786-O Human Renal Cancer Cells. Antioxidants (Basel) 2019; 8:antiox8100490. [PMID: 31627290 PMCID: PMC6826590 DOI: 10.3390/antiox8100490] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/08/2019] [Accepted: 10/12/2019] [Indexed: 12/28/2022] Open
Abstract
Clear-cell renal carcinoma (ccRCC) is the most common type of renal cancer. The importance of oxidative stress in the context of this disease has been described, although there is only little information concerning the role of superoxide dismutase (SOD) enzymes. The importance of SOD in different pathological conditions promoted the development of SOD mimics (SODm). As such, manganese(III) porphyrins can mimic the natural SOD enzymes and scavenge different reactive oxygen species (ROS), thus modulating the cellular redox status. In this study, the exposure of 786-O human renal cancer cells to MnTnHex-2-PyP5+ (MnP), a very promising SODm, led to a concentration and time-dependent decrease in cell viability and in the cell proliferation indices, as well as to an increase in apoptosis. No relevant effects in terms of micronuclei formation were observed. Moreover, the exposure to MnP resulted in a concentration-dependent increase in intracellular ROS, presumably due to the generation of H2O2 by the inherent redox mechanisms of MnP, along with the limited ability of cancer cells to detoxify this species. Although the MnP treatment did not result in a reduction in the collective cell migration, a significant decrease in chemotactic migration was observed. Overall, these results suggest that MnP has a beneficial impact on reducing renal cancer cell viability and migration and warrant further studies regarding SODm-based therapeutic strategies against human renal cancer.
Collapse
Affiliation(s)
- João G Costa
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal.
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Nuno Saraiva
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal.
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Ana S Fernandes
- Research Center for Biosciences & Health Technologies (CBIOS), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal.
| |
Collapse
|
14
|
Bharati SL, Sarma C, Hazarika PJ, Chaurasia PK, Anand N, Yadava S. Novel Mn(III) Porphyrins and Prospects of Their Application in Catalysis. RUSS J INORG CHEM+ 2019. [DOI: 10.1134/s0036023619030045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
Batinic-Haberle I, Tovmasyan A, Spasojevic I. Mn Porphyrin-Based Redox-Active Drugs: Differential Effects as Cancer Therapeutics and Protectors of Normal Tissue Against Oxidative Injury. Antioxid Redox Signal 2018; 29:1691-1724. [PMID: 29926755 PMCID: PMC6207162 DOI: 10.1089/ars.2017.7453] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE After approximatelty three decades of research, two Mn(III) porphyrins (MnPs), MnTE-2-PyP5+ (BMX-010, AEOL10113) and MnTnBuOE-2-PyP5+ (BMX-001), have progressed to five clinical trials. In parallel, another similarly potent metal-based superoxide dismutase (SOD) mimic-Mn(II)pentaaza macrocycle, GC4419-has been tested in clinical trial on application, identical to that of MnTnBuOE-2-PyP5+-radioprotection of normal tissue in head and neck cancer patients. This clearly indicates that Mn complexes that target cellular redox environment have reached sufficient maturity for clinical applications. Recent Advances: While originally developed as SOD mimics, MnPs undergo intricate interactions with numerous redox-sensitive pathways, such as those involving nuclear factor κB (NF-κB) and nuclear factor E2-related factor 2 (Nrf2), thereby impacting cellular transcriptional activity. An increasing amount of data support the notion that MnP/H2O2/glutathione (GSH)-driven catalysis of S-glutathionylation of protein cysteine, associated with modification of protein function, is a major action of MnPs on molecular level. CRITICAL ISSUES Differential effects of MnPs on normal versus tumor cells/tissues, which support their translation into clinic, arise from differences in their accumulation and redox environment of such tissues. This in turn results in different yields of MnP-driven modifications of proteins. Thus far, direct evidence for such modification of NF-κB, mitogen-activated protein kinases (MAPK), phosphatases, Nrf2, and endogenous antioxidative defenses was provided in tumor, while indirect evidence shows the modification of NF-κB and Nrf2 translational activities by MnPs in normal tissue. FUTURE DIRECTIONS Studies that simultaneously explore differential effects in same animal are lacking, while they are essential for understanding of extremely intricate interactions of metal-based drugs with complex cellular networks of normal and cancer cells/tissues.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Artak Tovmasyan
- 1 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Ivan Spasojevic
- 2 Department of Medicine, Duke University School of Medicine , Durham, North Carolina.,3 PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke Cancer Institute , Durham, North Carolina
| |
Collapse
|
16
|
Flórido A, Saraiva N, Cerqueira S, Almeida N, Parsons M, Batinic-Haberle I, Miranda JP, Costa JG, Carrara G, Castro M, Oliveira NG, Fernandes AS. The manganese(III) porphyrin MnTnHex-2-PyP 5+ modulates intracellular ROS and breast cancer cell migration: Impact on doxorubicin-treated cells. Redox Biol 2018; 20:367-378. [PMID: 30408752 PMCID: PMC6222139 DOI: 10.1016/j.redox.2018.10.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/19/2018] [Accepted: 10/21/2018] [Indexed: 12/21/2022] Open
Abstract
Manganese(III) porphyrins (MnPs) are superoxide dismutase (SOD) mimics with demonstrated beneficial effects in cancer treatment in combination with chemo- and radiotherapy regimens. Despite the ongoing clinical trials, little is known about the effect of MnPs on metastasis, being therefore essential to understand how MnPs affect this process. In the present work, the impact of the MnP MnTnHex-2-PyP5+ in metastasis-related processes was assessed in breast cancer cells (MCF-7 and MDA-MB-231), alone or in combination with doxorubicin (dox). The co-treatment of cells with non-cytotoxic concentrations of MnP and dox altered intracellular ROS, increasing H2O2. While MnP alone did not modify cell migration, the co-exposure led to a reduction in collective cell migration and chemotaxis. In addition, the MnP reduced the dox-induced increase in random migration of MDA-MB-231 cells. Treatment with either MnP or dox decreased the proteolytic invasion of MDA-MB-231 cells, although the effect was more pronounced upon co-exposure with both compounds. Moreover, to explore the cellular mechanisms underlying the observed effects, cell adhesion, spreading, focal adhesions, and NF-κB activation were also studied. Although differential effects were observed according to the endpoints analysed, overall, the alterations induced by MnP in dox-treated cells were consistent with a therapeutically favorable outcome. MnPs are SOD mimics with potential therapeutic applications in cancer. The impact of an MnP on breast cancer metastasis-related processes was assessed. Treatment with MnP+dox decreased collective cell migration, chemotaxis and invasion. MnP also reduced the dox-induced increase in random migration of MDA-MB-231 cells. Combination of MnP with dox revealed therapeutically favorable effects.
Collapse
Affiliation(s)
- Ana Flórido
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal
| | - Sara Cerqueira
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal
| | - Nuno Almeida
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, London SE1 1UL, England, UK
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Joana P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - João G Costa
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal; Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Guia Carrara
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Matilde Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisboa 1749-024, Portugal.
| |
Collapse
|
17
|
Dubey P, Yadava S. Synthesis and characterization of a novel Mn(III)-(γ-diketone) complex with catalytic and antifungal activity. J COORD CHEM 2018. [DOI: 10.1080/00958972.2018.1517869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Priti Dubey
- Department of Chemistry, DDU Gorakhpur University, Gorakhpur, India
| | - Sudha Yadava
- Department of Chemistry, DDU Gorakhpur University, Gorakhpur, India
| |
Collapse
|
18
|
Carballal S, Valez V, Alvarez-Paggi D, Tovmasyan A, Batinic-Haberle I, Ferrer-Sueta G, Murgida DH, Radi R. Manganese porphyrin redox state in endothelial cells: Resonance Raman studies and implications for antioxidant protection towards peroxynitrite. Free Radic Biol Med 2018; 126:379-392. [PMID: 30144631 DOI: 10.1016/j.freeradbiomed.2018.08.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Abstract
Cationic manganese(III) ortho N-substituted pyridylporphyrins (MnP) act as efficient antioxidants catalyzing superoxide dismutation and accelerating peroxynitrite reduction. Importantly, MnP can reach mitochondria offering protection against reactive species in different animal models of disease. Although an LC-MS/MS-based method for MnP quantitation and subcellular distribution has been reported, a direct method capable of evaluating both the uptake and the redox state of MnP in living cells has not yet been developed. In the present work we applied resonance Raman (RR) spectroscopy to analyze the intracellular accumulation of two potent MnP-based lipophilic SOD mimics, MnTnBuOE-2-PyP5+ and MnTnHex-2-PyP5+ within endothelial cells. RR experiments with isolated mitochondria revealed that the reduction of Mn(III)P was affected by inhibitors of the electron transport chain, supporting the action of MnP as efficient redox active compounds in mitochondria. Indeed, RR spectra confirmed that MnP added in the Mn(III) state can be incorporated into the cells, readily reduced by intracellular components to the Mn(II) state and oxidized by peroxynitrite. To assess the combined impact of reactivity and bioavailability, we studied the kinetics of Mn(III)TnBuOE-2-PyP5+ with peroxynitrite and evaluated the cytoprotective capacity of MnP by exposing the endothelial cells to nitro-oxidative stress induced by peroxynitrite. We observed a preservation of normal mitochondrial function, attenuation of cell damage and prevention of apoptotic cell death. These data introduce a novel application of RR spectroscopy for the direct detection of MnP and their redox states inside living cells, and helps to rationalize their antioxidant capacity in biological systems.
Collapse
Affiliation(s)
- Sebastián Carballal
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Valeria Valez
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Damián Alvarez-Paggi
- Departamento de Química Inorgánica, Analítica y Química Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. 2, piso 1, C1428EHA Buenos Aires, Argentina
| | - Artak Tovmasyan
- 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
| | - Gerardo Ferrer-Sueta
- Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay; Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Daniel H Murgida
- Departamento de Química Inorgánica, Analítica y Química Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. 2, piso 1, C1428EHA Buenos Aires, Argentina
| | - Rafael Radi
- Departmento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
19
|
Zhang XR, Zhou WX, Zhang YX. Improvements in SOD mimic AEOL-10150, a potent broad-spectrum antioxidant. Mil Med Res 2018; 5:30. [PMID: 30185231 PMCID: PMC6125955 DOI: 10.1186/s40779-018-0176-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/06/2018] [Indexed: 01/07/2023] Open
Abstract
AEOL-10150 is a broad-spectrum metalloporphyrin superoxidase dismutase (SOD) mimic specifically designed to neutralize reactive oxygen and nitrogen species. Research has shown that AEOL-10150 is a potent medical countermeasure against national security threats including sulfur mustard (SM), nerve agent exposure and radiation pneumonitis following a radiological/nuclear incident sufficient to cause acute radiation syndrome (ARS). AEOL-10150 performed well in animal safety studies, and two completed phase 1 safety studies in patients demonstrated that the drug was safe and well tolerated, indicating that AEOL-10150 has potential as a new catalytic antioxidant drug. In this article, we review improvements in AEOL-10150 in preclinical pharmacodynamic studies, especially regarding anti-SM, chlorine gas and radiation exposure studies.
Collapse
Affiliation(s)
- Xiao-Rui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Wen-Xia Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yong-Xiang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
20
|
Cline JM, Dugan G, Bourland JD, Perry DL, Stitzel JD, Weaver AA, Jiang C, Tovmasyan A, Owzar K, Spasojevic I, Batinic-Haberle I, Vujaskovic Z. Post-Irradiation Treatment with a Superoxide Dismutase Mimic, MnTnHex-2-PyP 5+, Mitigates Radiation Injury in the Lungs of Non-Human Primates after Whole-Thorax Exposure to Ionizing Radiation. Antioxidants (Basel) 2018. [PMID: 29518913 PMCID: PMC5874526 DOI: 10.3390/antiox7030040] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Radiation injury to the lung is the result of acute and chronic free radical formation, and there are currently few effective means of mitigating such injury. Studies in rodents indicate that superoxide dismutase mimetics may be effective in this regard; however, studies in humans or large animals are lacking. We hypothesized that post-exposure treatment with the lipophilic mitochondrial superoxide dismutase mimetic, MnTnHex-2-PyP5+ (hexyl), would reduce radiation-induced pneumonitis and fibrosis in the lungs of nonhuman primates. Rhesus monkeys (Macaca mulatta) received 10 Gy whole thorax irradiation, 10 Gy + hexyl treatment, sham irradiation, or sham irradiation + hexyl. Hexyl was given twice daily, subcutaneously, at 0.05 mg/kg, for 2 months. Animals were monitored daily, and respiratory rates, pulse oximetry, hematology and serum chemistry panels were performed weekly. Computed tomography scans were performed at 0, 2, and 4 months after irradiation. Supportive fluid therapy, corticosteroids, analgesics, and antibiotics were given as needed. All animals were humanely euthanized 4.5 months after irradiation, and pathologic assessments were made. Multifocal, progressive lung lesions were seen at 2 and 4 months in both irradiated groups. Hexyl treatment delayed the onset of radiation-induced lung lesions, reduced elevations of respiratory rate, and reduced pathologic increases in lung weight. No adverse effects of hexyl treatment were found. These results demonstrate (1) development of a nonhuman primate model of radiation-induced lung injury, (2) a significant mitigating effect of hexyl treatment on lung pathology in this model, and (3) no evidence for toxicity of hexyl at the dose studied.
Collapse
Affiliation(s)
- John Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA.
| | - Greg Dugan
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA.
| | - John Daniel Bourland
- Department of Radiation Oncology, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA.
| | - Donna L Perry
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1040, USA.
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Frederick, MD 21702, USA.
| | - Joel D Stitzel
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Biotech Place, 575 N. Patterson Ave., Winston-Salem, NC 21701, USA.
| | - Ashley A Weaver
- Department of Biomedical Engineering, Wake Forest University School of Medicine, Biotech Place, 575 N. Patterson Ave., Winston-Salem, NC 21701, USA.
| | - Chen Jiang
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27708, USA.
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27708, USA.
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC 27708, USA.
| | - Ivan Spasojevic
- Department of Medicine Duke University Medical Center, Durham, NC 27708, USA.
- Duke Cancer Institute, Pharmaceutical Research Shared Resource, PK/PD Core Laboratory, Duke University Medical Center, Durham, NC 27708, USA.
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27708, USA.
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27708, USA.
- Department of Radiation Oncology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
21
|
Rajic Z, Tovmasyan A, de Santana OL, Peixoto IN, Spasojevic I, do Monte SA, Ventura E, Rebouças JS, Batinic-Haberle I. Challenges encountered during development of Mn porphyrin-based, potent redox-active drug and superoxide dismutase mimic, MnTnBuOE-2-PyP 5+, and its alkoxyalkyl analogues. J Inorg Biochem 2017; 169:50-60. [PMID: 28131001 DOI: 10.1016/j.jinorgbio.2017.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/21/2016] [Accepted: 01/03/2017] [Indexed: 11/28/2022]
Abstract
We disclose here the studies that preceded and guided the preparation of the metal-based, redox-active therapeutic Mn(III) meso-tetrakis(N-n-butoxyethylpyridyl)porphyrin, MnTnBuOE-2-PyP5+ (BMX-001), which is currently in Phase I/II Clinical Trials at Duke University (USA) as a radioprotector of normal tissues in cancer patients. N-substituted pyridylporphyrins are ligands for Mn(III) complexes that are among the most potent superoxide dismutase mimics thus far synthesized. To advance their design, thereby improving their physical and chemical properties and bioavailability/toxicity profiles, we undertook a systematic study on placing oxygen atoms into N-alkylpyridyl chains via alkoxyalkylation reaction. For the first time we show here the unforeseen structural rearrangement that happens during the alkoxyalkylation reaction by the corresponding tosylates. Comprehensive experimental and computational approaches were employed to solve the rearrangement mechanism involved in quaternization of pyridyl nitrogens, which, instead of a single product, led to a variety of mixed N-alkoxyalkylated and N-alkylated pyridylporphyrins. The rearrangement mechanism involves the formation of an intermediate alkyl oxonium cation in a chain-length-dependent manner, which subsequently drives differential kinetics and thermodynamics of competing N-alkoxyalkylation versus in situ N-alkylation. The use of alkoxyalkyl tosylates, of different length of alkyl fragments adjacent to oxygen atom, allowed us to identify the set of alkyl fragments that would result in the synthesis of a single compound of high purity and excellent therapeutic potential.
Collapse
Affiliation(s)
- 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
| | - Otávio L de Santana
- Departamento de Química, CCEN, Universidade Federal da Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Isabelle N Peixoto
- Departamento de Química, CCEN, Universidade Federal da Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Duke Cancer Institute, Pharmaceutical Research Shared Resource, PK/PD Core Laboratory, Durham, NC 27710, United States
| | - Silmar A do Monte
- Departamento de Química, CCEN, Universidade Federal da Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Elizete Ventura
- Departamento de Química, CCEN, Universidade Federal da Paraiba, João Pessoa, PB 58051-900, Brazil
| | - Júlio S Rebouças
- Departamento de Química, CCEN, Universidade Federal da Paraiba, João Pessoa, PB 58051-900, Brazil.
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Moghnie S, Tovmasyan A, Craik J, Batinic-Haberle I, Benov L. Cationic amphiphilic Zn-porphyrin with high antifungal photodynamic potency. Photochem Photobiol Sci 2017; 16:1709-1716. [DOI: 10.1039/c7pp00143f] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Amphiphilic, cationic, water soluble Zn-porphyrins with six-carbon aliphatic chains attached to four pyridyl nitrogens atmesopositions of the porphyrin ring displayed high antifungal photo-efficiency and minimal dark toxicity.
Collapse
Affiliation(s)
- Sara Moghnie
- Department of Biochemistry
- Faculty of Medicine
- Kuwait University
- Safat 13110
- Kuwait
| | - Artak Tovmasyan
- Department of Radiation Oncology
- Duke University Medical Center
- Durham
- USA
| | - James Craik
- Department of Biochemistry
- Faculty of Medicine
- Kuwait University
- Safat 13110
- Kuwait
| | | | - Ludmil Benov
- Department of Biochemistry
- Faculty of Medicine
- Kuwait University
- Safat 13110
- Kuwait
| |
Collapse
|
23
|
Bharati SL, Chaurasia PK, Yadava S. Some novel organometallic MnIII complexes with porphine and 1,6-diaminohexane. RUSS J INORG CHEM+ 2016. [DOI: 10.1134/s0036023616020212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
24
|
Ochratoxin A-induced cytotoxicity, genotoxicity and reactive oxygen species in kidney cells: An integrative approach of complementary endpoints. Food Chem Toxicol 2016; 87:65-76. [DOI: 10.1016/j.fct.2015.11.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/19/2015] [Accepted: 11/21/2015] [Indexed: 01/26/2023]
|
25
|
Mn Porphyrin-Based Redox-Active Therapeutics. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2016. [DOI: 10.1007/978-3-319-30705-3_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Cui Y, Zeng L, Fang Y, Zhu J, Xu HJ, Desbois N, Gros CP, Kadish KM. Electrochemical and Spectroelectrochemical Properties of Free-Base Pyridyl- andN-Alkyl-4-Pyridylporphyrins in Nonaqueous Media. ChemElectroChem 2015. [DOI: 10.1002/celc.201500374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yan Cui
- Department of Chemistry; University of Houston; Houston TX 77204-5003 USA
| | - Lihan Zeng
- Department of Chemistry; University of Houston; Houston TX 77204-5003 USA
| | - Yuanyuan Fang
- Department of Chemistry; University of Houston; Houston TX 77204-5003 USA
| | - Jialiang Zhu
- Department of Chemistry; University of Houston; Houston TX 77204-5003 USA
| | - Hai-Jun Xu
- Université de Bourgogne; ICMUB UMR6302, CNRS, 9; Avenue Alain Savary, BP 47870 21078 Dijon Cedex France
- College of Chemical Engineering; Nanjing Forestry University; Nanjing 210037 China
| | - Nicolas Desbois
- Université de Bourgogne; ICMUB UMR6302, CNRS, 9; Avenue Alain Savary, BP 47870 21078 Dijon Cedex France
| | - Claude P. Gros
- Université de Bourgogne; ICMUB UMR6302, CNRS, 9; Avenue Alain Savary, BP 47870 21078 Dijon Cedex France
| | - Karl M. Kadish
- Department of Chemistry; University of Houston; Houston TX 77204-5003 USA
| |
Collapse
|
27
|
Tovmasyan A, Maia CGC, Weitner T, Carballal S, Sampaio RS, Lieb D, Ghazaryan R, Ivanovic-Burmazovic I, Ferrer-Sueta G, Radi R, Reboucas JS, Spasojevic I, Benov L, Batinic-Haberle I. A comprehensive evaluation of catalase-like activity of different classes of redox-active therapeutics. Free Radic Biol Med 2015; 86:308-21. [PMID: 26026699 PMCID: PMC4554972 DOI: 10.1016/j.freeradbiomed.2015.05.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 05/12/2015] [Accepted: 05/12/2015] [Indexed: 01/20/2023]
Abstract
Because of the increased insight into the biological role of hydrogen peroxide (H2O2) under physiological and pathological conditions and the role it presumably plays in the action of natural and synthetic redox-active drugs, there is a need to accurately define the type and magnitude of reactions that may occur with this intriguing and key species of redoxome. Historically, and frequently incorrectly, the impact of catalase-like activity has been assigned to play a major role in the action of many redox-active drugs, mostly SOD mimics and peroxynitrite scavengers, and in particular MnTBAP(3-) and Mn salen derivatives. The advantage of one redox-active compound over another has often been assigned to the differences in catalase-like activity. Our studies provide substantial evidence that Mn(III) N-alkylpyridylporphyrins couple with H2O2 in actions other than catalase-related. Herein we have assessed the catalase-like activities of different classes of compounds: Mn porphyrins (MnPs), Fe porphyrins (FePs), Mn(III) salen (EUK-8), and Mn(II) cyclic polyamines (SOD-active M40403 and SOD-inactive M40404). Nitroxide (tempol), nitrone (NXY-059), ebselen, and MnCl2, which have not been reported as catalase mimics, were used as negative controls, while catalase enzyme was a positive control. The dismutation of H2O2 to O2 and H2O was followed via measuring oxygen evolved with a Clark oxygen electrode at 25°C. The catalase enzyme was found to have kcat(H2O2)=1.5×10(6)M(-1) s(-1). The yield of dismutation, i.e., the maximal amount of O2 evolved, was assessed also. The magnitude of the yield reflects an interplay between the kcat(H2O2) and the stability of compounds toward H2O2-driven oxidative degradation, and is thus an accurate measure of the efficacy of a catalyst. The kcat(H2O2) values for 12 cationic Mn(III) N-substituted (alkyl and alkoxyalkyl) pyridylporphyrin-based SOD mimics and Mn(III) N,N'-dialkylimidazolium porphyrin, MnTDE-2-ImP(5+), ranged from 23 to 88M(-1) s(-1). The analogous Fe(III) N-alkylpyridylporphyrins showed ~10-fold higher activity than the corresponding MnPs, but the values of kcat(H2O2) are still ~4 orders of magnitude lower than that of the enzyme. While the kcat(H2O2) values for Fe ethyl and n-octyl analogs were 803.5 and 368.4M(-1) s(-1), respectively, the FePs are more prone to H2O2-driven oxidative degradation, therefore allowing for similar yields in H2O2 dismutation as analogous MnPs. The kcat(H2O2) values are dependent on the electron deficiency of the metal site as it controls the peroxide binding in the first step of the dismutation process. SOD-like activities depend on electron deficiency of the metal site also, as it controls the first step of O2(●-) dismutation. In turn, the kcat(O2(●-)) parallels the kcat(H2O2). Therefore, the electron-rich anionic non-SOD mimic MnTBAP(3-) has essentially very low catalase-like activity, kcat(H2O2)=5.8M(-1) s(-1). The catalase-like activities of Mn(III) and Fe(III) porphyrins are at most, 0.0004 and 0.05% of the enzyme activity, respectively. The kcat(H2O2) values of 8.2 and 6.5M(-1) s(-1) were determined for electron-rich Mn(II) cyclic polyamine-based compounds, M40403 and M40404, respectively. The EUK-8, with modest SOD-like activity, has only slightly higher kcat(H2O2)=13.5M(-1) s(-1). The biological relevance of kcat(H2O2) of MnTE-2-PyP(5+), MnTDE-2-ImP(5+), MnTBAP(3-), FeTE-2-PyP(5+), M40403, M40404, and Mn salen was evaluated in wild-type and peroxidase/catalase-deficient E. coli.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Clarissa G C Maia
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA; Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Tin Weitner
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sebastián Carballal
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Romulo S Sampaio
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA; Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Dominik Lieb
- Friedrich-Alexander Universitat, Erlangen-Nurnberg, Germany
| | - Robert Ghazaryan
- Department of Organic Chemistry, Faculty of Pharmacy, Yerevan State Medical University, Armenia
| | | | - Gerardo Ferrer-Sueta
- Laboratorio de Fisicoquímica Biológica, Facultad de Ciencias and Center for Free Radical and Biomedical Research, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Julio S Reboucas
- Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Pharmaceutical Research Shared Resource, PK/PD Core Laboratory, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
28
|
Slosky LM, Vanderah TW. Therapeutic potential of peroxynitrite decomposition catalysts: a patent review. Expert Opin Ther Pat 2015; 25:443-66. [PMID: 25576197 DOI: 10.1517/13543776.2014.1000862] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Peroxynitrite is a cytotoxic oxidant species implicated in a host of pathologies, including inflammatory and neurodegenerative diseases, cancer, radiation injury and chronic pain. With the recognition of the role of peroxynitrite in disease, numerous experimental and therapeutic tools have arisen to probe peroxyntirite's pathophysiological contribution and attenuate its oxidative damage. Peroxynitrite decomposition catalysts (PNDCs) are redox-active compounds that detoxify peroxynitrite by catalyzing its isomerization or reduction to nitrate or nitrite. AREAS COVERED This review discusses recent research articles and patents published 1995 - 2014 on the development and therapeutic use of PNDCs. Iron and manganese metalloporphyrin PNDCs attenuate the toxic effects of peroxynitrite and are currently being developed for clinical applications. Additionally, some Mn porphyrin-based PNDCs have optimized pharmaceutical properties such that they exhibit greater peroxynitrite selectivity. Other classes of PNDC agents, including bis(hydroxyphenyl)dipyrromethenes and metallocorroles, have demonstrated preclinical efficacy, oral availability and reduced toxicity risk. EXPERT OPINION Interest in the drug-like properties of peroxynitrite-neutralizing agents has grown with the realization that PNDCs will be powerful tools in the treatment of disease. The design of compounds with enhanced oral availability and peroxynitrite selectivity is a critical step toward the availability of safe, effective and selective redox modulators for the treatment of peroxynitrite-associated pathologies.
Collapse
Affiliation(s)
- Lauren M Slosky
- University of Arizona, Department of Pharmacology , Life Science North Rm 621, 1501 North Campbell Ave., Tucson, AZ 85721 , USA
| | | |
Collapse
|
29
|
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
|
30
|
Tovmasyan A, Carballal S, Ghazaryan R, Melikyan L, Weitner T, Maia CC, Reboucas JS, Radi R, Spasojevic I, Benov L, Batinic-Haberle I. Rational design of superoxide dismutase (SOD) mimics: the evaluation of the therapeutic potential of new cationic Mn porphyrins with linear and cyclic substituents. Inorg Chem 2014; 53:11467-83. [PMID: 25333724 PMCID: PMC4220860 DOI: 10.1021/ic501329p] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Indexed: 02/06/2023]
Abstract
Our goal herein has been to gain further insight into the parameters which control porphyrin therapeutic potential. Mn porphyrins (MnTnOct-2-PyP(5+), MnTnHexOE-2-PyP(5+), MnTE-2-PyPhP(5+), and MnTPhE-2-PyP(5+)) that bear the same positive charge and same number of carbon atoms at meso positions of porphyrin core were explored. The carbon atoms of their meso substituents are organized to form either linear or cyclic structures of vastly different redox properties, bulkiness, and lipophilicities. These Mn porphyrins were compared to frequently studied compounds, MnTE-2-PyP(5+), MnTE-3-PyP(5+), and MnTBAP(3-). All Mn(III) porphyrins (MnPs) have metal-centered reduction potential, E1/2 for Mn(III)P/Mn(II)P redox couple, ranging from -194 to +340 mV versus NHE, log kcat(O2(•-)) from 3.16 to 7.92, and log kred(ONOO(-)) from 5.02 to 7.53. The lipophilicity, expressed as partition between n-octanol and water, log POW, was in the range -1.67 to -7.67. The therapeutic potential of MnPs was assessed via: (i) in vitro ability to prevent spontaneous lipid peroxidation in rat brain homogenate as assessed by malondialdehyde levels; (ii) in vivo O2(•-) specific assay to measure the efficacy in protecting the aerobic growth of SOD-deficient Saccharomyces cerevisiae; and (iii) aqueous solution chemistry to measure the reactivity toward major in vivo endogenous antioxidant, ascorbate. Under the conditions of lipid peroxidation assay, the transport across the cellular membranes, and in turn shape and size of molecule, played no significant role. Those MnPs of E1/2 ∼ +300 mV were the most efficacious, significantly inhibiting lipid peroxidation in 0.5-10 μM range. At up to 200 μM, MnTBAP(3-) (E1/2 = -194 mV vs NHE) failed to inhibit lipid peroxidation, while MnTE-2-PyPhP(5+) with 129 mV more positive E1/2 (-65 mV vs NHE) was fully efficacious at 50 μM. The E1/2 of Mn(III)P/Mn(II)P redox couple is proportional to the log kcat(O2(•-)), i.e., the SOD-like activity of MnPs. It is further proportional to kred(ONOO(-)) and the ability of MnPs to prevent lipid peroxidation. In turn, the inhibition of lipid peroxidation by MnPs is also proportional to their SOD-like activity. In an in vivo S. cerevisiae assay, however, while E1/2 predominates, lipophilicity significantly affects the efficacy of MnPs. MnPs of similar log POW and E1/2, that have linear alkyl or alkoxyalkyl pyridyl substituents, distribute more easily within a cell and in turn provide higher protection to S. cerevisiae in comparison to MnP with bulky cyclic substituents. The bell-shape curve, with MnTE-2-PyP(5+) exhibiting the highest ability to catalyze ascorbate oxidation, has been established and discussed. Our data support the notion that the SOD-like activity of MnPs parallels their therapeutic potential, though species other than O2(•-), such as peroxynitrite, H2O2, lipid reactive species, and cellular reductants, may be involved in their mode(s) of action(s).
Collapse
Affiliation(s)
- Artak Tovmasyan
- Departments of Radiation Oncology and Medicine, Duke University Medical Center, Research Drive, 281b MSRB I, Durham, North Carolina 27710, United States
| | - Sebastian Carballal
- Departamento
de Bioquímica and Center for Free Radical and Biomedical
Research, Facultad de Medicina, Universidad
de la República, Montevideo, Uruguay
| | - Robert Ghazaryan
- Department of Organic Chemistry, Faculty
of Pharmacy, Yerevan State Medical University, Yerevan, Armenia
| | - Lida Melikyan
- Department of Organic Chemistry, Faculty
of Pharmacy, Yerevan State Medical University, Yerevan, Armenia
| | - Tin Weitner
- Departments of Radiation Oncology and Medicine, Duke University Medical Center, Research Drive, 281b MSRB I, Durham, North Carolina 27710, United States
| | - Clarissa
G. C. Maia
- Departamento de Quimica, CCEN, Universidade
Federal de Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Julio S. Reboucas
- Departamento de Quimica, CCEN, Universidade
Federal de Paraiba, Joao Pessoa, PB 58051-900, Brazil
| | - Rafael Radi
- Departamento
de Bioquímica and Center for Free Radical and Biomedical
Research, Facultad de Medicina, Universidad
de la República, Montevideo, Uruguay
| | - Ivan Spasojevic
- Departments of Radiation Oncology and Medicine, Duke University Medical Center, Research Drive, 281b MSRB I, Durham, North Carolina 27710, United States
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Ines Batinic-Haberle
- Departments of Radiation Oncology and Medicine, Duke University Medical Center, Research Drive, 281b MSRB I, Durham, North Carolina 27710, United States
| |
Collapse
|
31
|
Celic T, Španjol J, Bobinac M, Tovmasyan A, Vukelic I, Reboucas JS, Batinic-Haberle I, Bobinac D. Mn porphyrin-based SOD mimic, MnTnHex-2-PyP(5+), and non-SOD mimic, MnTBAP(3-), suppressed rat spinal cord ischemia/reperfusion injury via NF-κB pathways. Free Radic Res 2014; 48:1426-42. [PMID: 25185063 DOI: 10.3109/10715762.2014.960865] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Herein we have demonstrated that both superoxide dismutase (SOD) mimic, cationic Mn(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTnHex-2-PyP(5+)), and non-SOD mimic, anionic Mn(III) meso-tetrakis(4-carboxylatophenyl)porphyrin (MnTBAP(3-)), protect against oxidative stress caused by spinal cord ischemia/reperfusion via suppression of nuclear factor kappa B (NF-κB) pro-inflammatory pathways. Earlier reports showed that Mn(III) N-alkylpyridylporphyrins were able to prevent the DNA binding of NF-κB in an aqueous system, whereas MnTBAP(3-) was not. Here, for the first time, in a complex in vivo system-animal model of spinal cord injury-a similar impact of MnTBAP(3-), at a dose identical to that of MnTnHex-2-PyP(5+), was demonstrated in NF-κB downregulation. Rats were treated subcutaneously at 1.5 mg/kg starting at 30 min before ischemia/reperfusion, and then every 12 h afterward for either 48 h or 7 days. The anti-inflammatory effects of both Mn porphyrins (MnPs) were demonstrated in the spinal cord tissue at both 48 h and 7 days. The downregulation of NF-κB, a major pro-inflammatory signaling protein regulating astrocyte activation, was detected and found to correlate well with the suppression of astrogliosis (as glial fibrillary acidic protein) by both MnPs. The markers of oxidative stress, lipid peroxidation and protein carbonyl formation, were significantly reduced by MnPs. The favorable impact of both MnPs on motor neurons (Tarlov score and inclined plane test) was assessed. No major changes in glutathione peroxidase- and SOD-like activities were demonstrated, which implies that none of the MnPs acted as SOD mimic. Increasing amount of data on the reactivity of MnTBAP(3-) with reactive nitrogen species (RNS) (.NO/HNO/ONOO(-)) suggests that RNS/MnTBAP(3-)-driven modification of NF-κB protein cysteines may be involved in its therapeutic effects. This differs from the therapeutic efficacy of MnTnHex-2-PyP(5+) which presumably occurs via reactive oxygen species and relates to NF-κB thiol oxidation; the role of RNS cannot be excluded.
Collapse
Affiliation(s)
- T Celic
- Department of Anatomy, Faculty of Medicine, University of Rijeka , Rijeka , Croatia
| | | | | | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Sheng H, Chaparro RE, Sasaki T, Izutsu M, Pearlstein RD, Tovmasyan A, Warner DS. Metalloporphyrins as therapeutic catalytic oxidoreductants in central nervous system disorders. Antioxid Redox Signal 2014; 20:2437-64. [PMID: 23706004 DOI: 10.1089/ars.2013.5413] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Metalloporphyrins, characterized by a redox-active transitional metal (Mn or Fe) coordinated to a cyclic porphyrin core ligand, mitigate oxidative/nitrosative stress in biological systems. Side-chain substitutions tune redox properties of metalloporphyrins to act as potent superoxide dismutase mimics, peroxynitrite decomposition catalysts, and redox regulators of transcription factor function. With oxidative/nitrosative stress central to pathogenesis of CNS injury, metalloporphyrins offer unique pharmacologic activity to improve the course of disease. RECENT ADVANCES Metalloporphyrins are efficacious in models of amyotrophic lateral sclerosis, Alzheimer's disease, epilepsy, neuropathic pain, opioid tolerance, Parkinson's disease, spinal cord injury, and stroke and have proved to be useful tools in defining roles of superoxide, nitric oxide, and peroxynitrite in disease progression. The most substantive recent advance has been the synthesis of lipophilic metalloporphyrins offering improved blood-brain barrier penetration to allow intravenous, subcutaneous, or oral treatment. CRITICAL ISSUES Insufficient preclinical data have accumulated to enable clinical development of metalloporphyrins for any single indication. An improved definition of mechanisms of action will facilitate preclinical modeling to define and validate optimal dosing strategies to enable appropriate clinical trial design. Due to previous failures of "antioxidants" in clinical trials, with most having markedly less biologic activity and bioavailability than current-generation metalloporphyrins, a stigma against antioxidants has discouraged the development of metalloporphyrins as CNS therapeutics, despite the consistent definition of efficacy in a wide array of CNS disorders. FUTURE DIRECTIONS Further definition of the metalloporphyrin mechanism of action, side-by-side comparison with "failed" antioxidants, and intense effort to optimize therapeutic dosing strategies are required to inform and encourage clinical trial design.
Collapse
Affiliation(s)
- Huaxin Sheng
- 1 Department of Anesthesiology, Duke University Medical Center (DUMC) , Durham, North Carolina
| | | | | | | | | | | | | |
Collapse
|
34
|
Ramos CIV, Pereira PMR, Santana-Marques MG, De Paula R, Simões MMQ, Neves MGPMS, Cavaleiro JAS. Imidazole and imidazolium porphyrins: gas-phase chemistry of multicharged ions. JOURNAL OF MASS SPECTROMETRY : JMS 2014; 49:371-379. [PMID: 24809898 DOI: 10.1002/jms.3350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 02/12/2014] [Accepted: 02/16/2014] [Indexed: 06/03/2023]
Abstract
Electrospray ionization mass spectrometry/mass spectrometry in the positive ion mode was used to investigate the gas-phase chemistry of multicharged ions from solutions of porphyrins with 1,3-dimethylimidazolium-2-yl (DMIM) and 1-methylimidazol-2-yl (MIm) meso-substituents. The studied compounds include two free bases and 12 complexes with transition metals (Cu(II), Zn(II), Mn(III), and Fe(III)). The observed multicharged ions are either preformed or formed during the electrospraying process by reduction or protonation and comprise closed-shell and hypervalent mono-radical and bi-radical ions. The observed extensive and abundant fragmentation of the DMIM and MIm meso-substituents is a characteristic feature of these porphyrins. Fragments with the same mass values can be lost from the meso-substituents either as charged or neutral species and from closed-shell and hypervalent radical ions. Reduction processes are observed for both the free bases and the metallated DMIM porphyrins and occur predominantly by formation of hypervalent radicals that fragment, at low energy collisions, by loss of methyl radicals with formation of the corresponding MIm functionalities. These findings confirm that, when using electrospray ionization, reduction is an important characteristic of cationic meso-substituted tetrapyrrolic macrocycles, always occurring when delocalization of the formed hypervalent radicals is possible. For the Fe(III) and Mn(III) complexes, reduction of the metal centers is also observed as the predominant fragmentation of the corresponding reduced ions through losses of charged fragments testifies. The fragmentation of the closed-shell ions formed by protonation of the MIm porphyrins mirrors the fragmentation of the closed-shell ions of their DMIM counterparts.
Collapse
Affiliation(s)
- Catarina I V Ramos
- Mass Spectrometry Laboratory, QOPNA, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | | | | | | | | | | | | |
Collapse
|
35
|
Gauter-Fleckenstein B, Reboucas JS, Fleckenstein K, Tovmasyan A, Owzar K, Jiang C, Batinic-Haberle I, Vujaskovic Z. Robust rat pulmonary radioprotection by a lipophilic Mn N-alkylpyridylporphyrin, MnTnHex-2-PyP(5+). Redox Biol 2014; 2:400-10. [PMID: 24624330 PMCID: PMC3949096 DOI: 10.1016/j.redox.2013.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
With the goal to enhance the distribution of cationic Mn porphyrins within mitochondria, the lipophilic Mn(III)meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin, MnTnHex-2-PyP(5+) has been synthesized and tested in several different model of diseases, where it shows remarkable efficacy at as low as 50 µg/kg single or multiple doses. Yet, in a rat lung radioprotection study, at higher 0.6-1 mg/kg doses, due to its high accumulation and micellar character, it became toxic. To avoid the toxicity, herein the pulmonary radioprotection of MnTnHex-2-PyP(5+) was assessed at 50 µg/kg. Fischer rats were irradiated to their right hemithorax (28 Gy) and treated with 0.05 mg/kg/day of MnTnHex-2-PyP(5+) for 2 weeks by subcutaneously-implanted osmotic pumps, starting at 2 h post-radiation. The body weights and breathing frequencies were followed for 10 weeks post-radiation, when the histopathology and immunohistochemistry were assessed. Impact of MnTnHex-2-PyP(5+) on macrophage recruitment (ED-1), DNA oxidative damage (8-OHdG), TGF-β1, VEGF(A) and HIF-1α were measured. MnTnHex-2-PyP(5+) significantly decreased radiation-induced lung histopathological (H&E staining) and functional damage (breathing frequencies), suppressed oxidative stress directly (8-OHdG), or indirectly, affecting TGF-β1, VEGF (A) and HIF-1α pathways. The magnitude of the therapeutic effects is similar to the effects demonstrated under same experimental conditions with 120-fold higher dose of ~5000-fold less lipophilic Mn(III)meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+).
Collapse
Key Words
- 8-OHdG, 8-hydroxy-2'-deoxyguanosine
- AKT, protein kinase B (PKB), a serine/threonine-specific protein kinase
- ALS, amyotrophic laterial sclerosis
- AP-1, activator protein-1
- AT, ataxia telangiectasia
- BBB, blood brain barrier
- Breathing frequencies
- CNS, central nervous system
- CO3−, carbonate radical
- ClO−, hypochlorite
- ETC, mitochondrial electron transport chain
- Fischer rats
- GMP, good manufacturing practice
- GS−, monodeprotonated glutathione
- HIF-1α, hypoxia inducible factor-1
- HO2−, monodeprotonated hydrogen peroxide
- Histopathology
- I/R, ischemia reperfusion
- Immunohistochemistry
- Lung injury
- MCAO, middle cerebral artery occlusion
- Manganese porphyrins
- MnP, Mn porphyrin
- MnTDE-2-ImP5+, Mn(III) tetrakis[N,N'-diethylimidazolium-2-yl)porphyrin, AEOL10150
- MnTE-2-PyP5+
- MnTE-2-PyP5+, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (AEOL10113)
- MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis(N-(n-butoxyethyl)pyridinium-2-yl)porphyrin
- MnTnHex-2-PyP5+
- MnTnHex-2-PyP5+, Mn(III) meso-tetrakis(N-(n-hexyl)pyridinium-2-yl)porphyrin (AEOL10113)
- NF-κB, nuclear factor κB
- NHE, normal hydrogen electrode
- NO, nitric oxide
- NOX4, NADPH oxidase, isoform 4 E1/2, Half-wave metal-centered reduction potential
- Nrf-2, nuclear factor-erythroid-derived 2-like 2
- O2−, superoxide
- ONOO−, peroxynitrite
- PI3K, phosphatidylinositide 3-kinase
- PTEN, phosphoinositide 3-phosphatase
- Radioprotection
- Redox-modulators
- SAH, subarachnoid hemorrhage
- SOD, superoxide dismutase
- SP-1, specificity protein-1
- TF, transcription factor
- TGF-β1, one of the 3 members of the TGF-β transforming growth factor-β family
- VEGF, vascular endothelial growth factor
- mTOR, mammalian target of rapamycin (mTOR), a serine/threonine protein kinase
Collapse
Affiliation(s)
- Benjamin Gauter-Fleckenstein
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julio S Reboucas
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Katharina Fleckenstein
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, USA ; Biostatistics and Computational Biology Core, RadCCORE, Duke University Medical Center, Durham, USA
| | - Chen Jiang
- Biostatistics and Computational Biology Core, RadCCORE, Duke University Medical Center, Durham, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, 655W Baltimore Street, Bressler Research Building, 8-025, Baltimore, MD 21201, USA
| |
Collapse
|
36
|
Kanetada N, Matsumura C, Yamazaki S, Adachi K. Mechanism of peripheral substituent effects on adsorption-aggregation behaviors of cationic porphyrin dyes on tungsten(VI) oxide nanocolloid particles. ACS APPLIED MATERIALS & INTERFACES 2013; 5:12991-12999. [PMID: 24298877 DOI: 10.1021/am403751t] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The adsorption and aggregation behaviors of the cationic porphyrin derivatives such as 5,10,15,20-tetrakis(4-pyridyl)porphyrin [TPyP], 5,10,15,20-tetrakis(N-methyl-4-pyridyl)porphyrin [TMPyP], 5,10,15,20-tetrakis(N-ethyl-4-pyridyl)porphyrin [TEPyP], and 5,10,15,20-tetrakis(N-n-propyl-4-pyridyl)porphyrin [TPPyP] (hereafter called "TPyP derivatives") in the tungsten(VI) oxide (WO3) colloid aqueous solution at weak acidic pH were studied by UV-vis spectroscopy. The TPyP derivatives were strongly adsorbed as monolayer onto the WO3 surface via the electrostatic interaction between their peripheral cationic substituents and negatively surface-charged WO3 colloid particles, and most of the ones adsorbed eventually formed J-type dimers aligned in the head-to-tail fashion. These different dimerization states were effectively analyzed by the change of ratios among the intensities of exciton split Soret bands (H- and J-bands). Judging from the exciton coupling theory and adsorption measurements, we concluded that the J-dimer geometry of the TPyP derivatives adsorbed on the WO3 colloid particle surface is strongly dependent on the presence and difference of peripheral substituents. The results described here indicate a new and promising way of designing surface supramolecular structures combination of two principles, the self-association of organic dyes, and the steric repulsive interaction between the peripheral substituents and the inorganic semiconductor surfaces.
Collapse
Affiliation(s)
- Naoya Kanetada
- Department of Environmental Science & Engineering, Graduate School of Science & Engineering, Yamaguchi University , Yamaguchi 753-8512, Japan
| | | | | | | |
Collapse
|
37
|
Ezzeddine R, Al-Banaw A, Tovmasyan A, Craik JD, Batinic-Haberle I, Benov LT. Effect of molecular characteristics on cellular uptake, subcellular localization, and phototoxicity of Zn(II) N-alkylpyridylporphyrins. J Biol Chem 2013; 288:36579-88. [PMID: 24214973 DOI: 10.1074/jbc.m113.511642] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tetra-cationic Zn(II) meso-tetrakis(N-alkylpyridinium-2 (or -3 or -4)-yl)porphyrins (ZnPs) with progressively increased lipophilicity were synthesized to investigate how the tri-dimensional shape and lipophilicity of the photosensitizer (PS) affect cellular uptake, subcellular distribution, and photodynamic efficacy. The effect of the tri-dimensional shape of the molecule was studied by shifting the N-alkyl substituent attached to the pyridyl nitrogen from ortho to meta and para positions. Progressive increase of lipophilicity from shorter hydrophilic (methyl) to longer amphiphilic (hexyl) alkyl chains increased the phototoxicity of the ZnP PSs. PS efficacy was also increased for all derivatives when the alkyl substituents were shifted from ortho to meta, and from meta to para positions. Both cellular uptake and subcellular distribution of the PSs were affected by the lipophilicity and the position of the alkyl chains on the periphery of the porphyrin ring. Whereas the hydrophilic ZnPs demonstrated mostly lysosomal distribution, the amphiphilic hexyl derivatives were associated with mitochondria, endoplasmic reticulum, and plasma membrane. A comparison of hexyl isomers revealed that cellular uptake and partition into membranes followed the order para > meta > ortho. Varying the position and length of the alkyl substituents affects (i) the exposure of cationic charges for electrostatic interactions with anionic biomolecules and (ii) the lipophilicity of the molecule. The charge, lipophilicity, and the tri-dimensional shape of the PS are the major factors that determine cellular uptake, subcellular distribution, and as a consequence, the phototoxicity of the PSs.
Collapse
Affiliation(s)
- Rima Ezzeddine
- From the Department of Biochemistry, Faculty of Medicine, and
| | | | | | | | | | | |
Collapse
|
38
|
Weitner T, Kos I, Mandić Z, Batinić-Haberle I, Biruš M. Acid-base and electrochemical properties of manganese meso(ortho- and meta-N-ethylpyridyl)porphyrins: voltammetric and chronocoulometric study of protolytic and redox equilibria. Dalton Trans 2013; 42:14757-65. [PMID: 23933742 PMCID: PMC3876927 DOI: 10.1039/c3dt50767j] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Growing interest in redox-active compounds as therapeutics for oxidative stress-related diseases led to the design of metalloporphyrins as some of the most potent functional SOD-mimics. Herein we report a detailed electrochemical study of the protolytic and redox equilibria of manganese ortho and meta substituted N-ethylpyridyl porphyrins (MnPs), MnTE-2-PyP(5+) and MnTE-3-PyP(5+), in aqueous solutions. The electrochemical parameters of redox processes for all experimentally available species have been determined, as well as their diffusion coefficients and estimated sizes of aqueous cavities. The results indicate that possible changes of the intracellular acidity cannot affect the antioxidant activity of MnPs in vivo, since no change in the E(Mn(III)P/Mn(II)P) values was observed below pH 10. Furthermore, the results confirm that both of these MnPs can be efficient redox scavengers of peroxynitrite (ONOO(-)), another major damaging species in vivo. This can occur by either single-electron reduction or two-electron reduction of ONOO(-), involving either the Mn(IV)P/Mn(III)P redox couple or Mn(IV)P/Mn(II)P redox couple. In addition to kred(ONOO(-)) reported previously, the thermodynamic parameters calculated herein imply a strong and identical driving force for the reaction of both ortho and meta isomeric MnPs with ONOO(-). An enlargement of both Mn(III)P complexes upon an increase of the solution pH was also observed and attributed to the reduction of positive charge on the central ion caused by deprotonation of the axial water molecules. This expansion of aqueous cavities suggests the formation of a solvent cage and the increased lipophilicity of Mn(III)P complexes caused by increased electron density on the Mn ion.
Collapse
Affiliation(s)
- Tin Weitner
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, Zagreb 10000, Croatia.
| | | | | | | | | |
Collapse
|
39
|
Bioenergetic deficits in peripheral nerve sensory axons during chemotherapy-induced neuropathic pain resulting from peroxynitrite-mediated post-translational nitration of mitochondrial superoxide dismutase. Pain 2013; 154:2432-2440. [PMID: 23891899 DOI: 10.1016/j.pain.2013.07.032] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/19/2013] [Accepted: 07/17/2013] [Indexed: 11/23/2022]
Abstract
Many of the widely used anticancer drugs induce dose-limiting peripheral neuropathies that undermine their therapeutic efficacy. Animal models of chemotherapy-induced painful peripheral neuropathy (CIPN) evoked by a variety of drug classes, including taxanes, vinca alkaloids, platinum-complexes, and proteasome-inhibitors, suggest that the common underlying mechanism in the development of these neuropathies is mitotoxicity in primary nerve sensory axons (PNSAs) arising from reduced mitochondrial bioenergetics [eg adenosine triphosphate (ATP) production deficits due to compromised respiratory complex I and II activity]. The causative mechanisms of this mitotoxicity remain poorly defined. However, peroxynitrite, an important pro-nociceptive agent, has been linked to mitotoxicity in several disease states and may also drive the mitotoxicity associated with CIPN. Our findings reveal that the development of mechano-hypersensitivity induced by paclitaxel, oxaliplatin, and bortezomib was prevented by administration of the peroxynitrite decomposition catalyst Mn(III) 5,10,15,20-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) without interfering with their anti-tumor effects. Peak CIPN was associated with the nitration and inactivation of superoxide dismutase in the mitochondria, but not in the cytosol, as well as a significant decrease in ATP production within the PNSAs; all of these events were attenuated by MnTE-2-PyP(5+). Our results provide continued support for the role of mitotoxicity in the development of CIPN across chemotherapeutic drug classes, and identify peroxynitrite as a key mediator in these processes, thereby providing the rationale towards development of "peroxynitrite-targeted" therapeutics for CIPN.
Collapse
|
40
|
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
|
41
|
Aitken JB, Shearer EL, Giles NM, Lai B, Vogt S, Reboucas JS, Batinic-Haberle I, Lay PA, Giles GI. Intracellular Targeting and Pharmacological Activity of the Superoxide Dismutase Mimics MnTE-2-PyP5+ and MnTnHex-2-PyP5+ Regulated by Their Porphyrin Ring Substituents. Inorg Chem 2013; 52:4121-3. [DOI: 10.1021/ic300700g] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jade B. Aitken
- School of
Chemistry, The University of Sydney, NSW
2006, Australia
| | - Emily L. Shearer
- Department
of Pharmacology and Toxicology, University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Niroshini M. Giles
- Department
of Pharmacology and Toxicology, University of Otago, P.O. Box 913, Dunedin, New Zealand
| | - Barry Lai
- X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Stefan Vogt
- X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Julio S. Reboucas
- Departamento de Quimica, Universidade Federal da Paraiba, Joao Pessoa, PB, Brazil
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University, Durham, North Carolina 27710, United
States
| | - Peter A. Lay
- School of
Chemistry, The University of Sydney, NSW
2006, Australia
| | - Gregory I. Giles
- Department
of Pharmacology and Toxicology, University of Otago, P.O. Box 913, Dunedin, New Zealand
| |
Collapse
|
42
|
Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, Neumann WL, Salvemini D. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain 2013; 154:978-86. [PMID: 23590939 DOI: 10.1016/j.pain.2013.02.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/15/2013] [Accepted: 02/21/2013] [Indexed: 10/27/2022]
Abstract
Treatment of severe pain by morphine, the gold-standard opioid and a potent drug in our arsenal of analgesic medications, is limited by the eventual development of hyperalgesia and analgesic tolerance. We recently reported that systemic administration of a peroxynitrite (PN) decomposition catalyst (PNDC) or superoxide dismutase mimetic attenuates morphine hyperalgesia and antinociceptive tolerance and reduces PN-mediated mitochondrial nitroxidative stress in the spinal cord. These results suggest the potential involvement of spinal PN signaling in this setting; which was examined in the present study. PN removal with intrathecal delivery of manganese porphyrin-based dual-activity superoxide/PNDCs, MnTE-2-PyP(5+) and the more lipophilic MnTnHex-2-PyP(5+), blocked hyperalgesia and antinociceptive tolerance in rats. Noteworthy is that intrathecal MnTnHex-2-PyP(5+) prevented nitration and inactivation of mitochondrial manganese superoxide dismutase. Mitochondrial manganese superoxide dismutase inactivation enhances the superoxide-to-PN pathway by preventing the dismutation of superoxide to hydrogen peroxide, thus providing an important enzymatic source for PN formation. Additionally, intrathecal MnTnHex-2-PyP(5+) attenuated neuroimmune activation by preventing the activation of nuclear factor kappa B, extracellular-signal-regulated kinase and p38 mitogen activated protein kinases, and the enhanced levels of proinflammatory cytokines, interleukin (IL)-1β and IL-6, while increasing anti-inflammatory cytokines, IL-4 and IL-10. The role of PN was further confirmed using intrathecal or oral delivery of the superoxide-sparing PNDC, SRI-110. These results suggest that mitochondrial-derived PN triggers the activation of several biochemical pathways engaged in the development of neuroinflammation in the spinal cord that are critical to morphine hyperalgesia and tolerance, further supporting the potential of targeting PN as an adjunct to opiates to maintain pain relief.
Collapse
|
43
|
Batinic-Haberle I, Tovmasyan A, Spasojevic I. The complex mechanistic aspects of redox-active compounds, commonly regarded as SOD mimics. ACTA ACUST UNITED AC 2013. [DOI: 10.1515/irm-2013-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
44
|
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
|
45
|
Mouraviev V, Venkatraman TN, Tovmasyan A, Kimura M, Tsivian M, Mouravieva V, Polascik TJ, Wang H, Amrhein TJ, Batinic-Haberle I, Lascola C. Mn porphyrins as novel molecular magnetic resonance imaging contrast agents. J Endourol 2012; 26:1420-4. [PMID: 22783812 DOI: 10.1089/end.2012.0171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND PURPOSE In this study, we investigated the potential of a new class of therapeutic Mn porphyrins as molecular MRI probes for prostate cancer imaging. Two compounds of different bioavailibility were investigated: Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) and Mn(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (MnTnHex-2-PyP(5+)). These compounds have previously been shown to have adjunctive antineoplastic activity through their actions as powerful superoxide dismutase mimics, peroxynitrite scavengers, and modulators of cellular redox-based signaling pathways. Strong paramagnetic MRI contrast properties and affinity for cancer cells suggest their potential application as novel diagnostic imaging agents. MATERIALS AND METHODS MRI experiments were performed at 7.0T on a Bruker Biospec horizontal bore scanner. All in-vivo experiments were performed on 12 C57 black mice implanted with RM-9 prostate cancer cells on the hind limb. Two mg/kg of MnTnHex-2-PyP(5+) (n=6) and 8 mg/kg MnTE-2-PyP(5+) (n=6) were administered intraperitoneally 90 minutes before imaging. All the images were collected using a volume coil and processed using Paravision 4.0. RESULTS Phantom studies reveal remarkably high T1 relaxivity changes for both metalloporphyrins, which are twofold to threefold higher than commercially available gadolinium chelates. Observable detection limits using conventional T1-weighted MRI are in the low micromolar range for both compounds. In vivo, MR relaxation changes in prostate tumor xenografts were readily observed after a single injection of either MnTE-2-PyP(5+)or MnTnHex-2-PyP(5+), with tumor contrast to background ratio greatest after MnTE-2-PyP(5+) administration. CONCLUSION After a single dose of MnTE-2-PyP(5+), contrast changes in prostate tumors are up to sixfold greater than in surrounding, noncancerous tissues, suggesting the potential use of this metalloporphyrin as a novel diagnostic probe for detecting prostate malignancy using MRI.
Collapse
Affiliation(s)
- Vladimir Mouraviev
- Department of Urology/Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bharati SL, Yadava S. Some Mn III-porphyrins with de-polymerization activity toward humic acid. J COORD CHEM 2012. [DOI: 10.1080/00958972.2012.718763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Shashi Lata Bharati
- a Department of Chemistry , D.D.U. Gorakhpur University , Gorakhpur – 273009 , India
| | - Sudha Yadava
- a Department of Chemistry , D.D.U. Gorakhpur University , Gorakhpur – 273009 , India
| |
Collapse
|
47
|
Rajic Z, Tovmasyan A, Spasojevic I, Sheng H, Lu M, Li AM, Gralla EB, Warner DS, Benov L, Batinic-Haberle I. A new SOD mimic, Mn(III) ortho N-butoxyethylpyridylporphyrin, combines superb potency and lipophilicity with low toxicity. Free Radic Biol Med 2012; 52:1828-34. [PMID: 22336516 PMCID: PMC3353805 DOI: 10.1016/j.freeradbiomed.2012.02.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/17/2012] [Accepted: 02/03/2012] [Indexed: 10/28/2022]
Abstract
The Mn porphyrins of k(cat)(O(2)(.-)) as high as that of a superoxide dismutase enzyme and of optimized lipophilicity have already been synthesized. Their exceptional in vivo potency is at least in part due to their ability to mimic the site and location of mitochondrial superoxide dismutase, MnSOD. MnTnHex-2-PyP(5+) is the most studied among lipophilic Mn porphyrins. It is of remarkable efficacy in animal models of oxidative stress injuries and particularly in central nervous system diseases. However, when used at high single and multiple doses it becomes toxic. The toxicity of MnTnHex-2-PyP(5+) has been in part attributed to its micellar properties, i.e., the presence of polar cationic nitrogens and hydrophobic alkyl chains. The replacement of a CH(2) group by an oxygen atom in each of the four alkyl chains was meant to disrupt the porphyrin micellar character. When such modification occurs at the end of long alkyl chains, the oxygens become heavily solvated, which leads to a significant drop in the lipophilicity of porphyrin. However, when the oxygen atoms are buried deeper within the long heptyl chains, their excessive solvation is precluded and the lipophilicity preserved. The presence of oxygens and the high lipophilicity bestow the exceptional chemical and physical properties to Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP(5+). The high SOD-like activity is preserved and even enhanced: log k(cat)(O(2)(.-))=7.83 vs 7.48 and 7.65 for MnTnHex-2-PyP(5+) and MnTnHep-2-PyP(5+), respectively. MnTnBuOE-2-PyP(5+) was tested in an O(2)(.-) -specific in vivo assay, aerobic growth of SOD-deficient yeast, Saccharomyces cerevisiae, where it was fully protective in the range of 5-30 μM. MnTnHep-2-PyP(5+) was already toxic at 5 μM, and MnTnHex-2-PyP(5+) became toxic at 30 μM. In a mouse toxicity study, MnTnBuOE-2-PyP(5+) was several-fold less toxic than either MnTnHex-2-PyP(5+) or MnTnHep-2-PyP(5+).
Collapse
Affiliation(s)
- Zrinka Rajic
- Department of Radiation Oncology, Duke University Medical Center, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, NC 27710, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, NC 27710, USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University Medical Center, NC 27710, USA
| | - Miaomiao Lu
- Department of Anesthesiology, Duke University Medical Center, NC 27710, USA
- Department of Anesthesiology, Second Affiliated Hospital, Zhengzhou University, Henan, China
| | - Alice M. Li
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095-1569, USA
| | - Edith B. Gralla
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095-1569, USA
| | - David S. Warner
- Department of Anesthesiology, Duke University Medical Center, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, 13110 Safat, Kuwait
| | | |
Collapse
|
48
|
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
|
49
|
Yadava S, Bharati SL. Novel complexes of Mn(III) with macrocylic porphine ligand and ethylenediamine. J COORD CHEM 2011. [DOI: 10.1080/00958972.2011.632412] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Sudha Yadava
- a Department of Chemistry , DDU Gorakhpur University , Gorakhpur-273009, India
| | - Shashi Lata Bharati
- a Department of Chemistry , DDU Gorakhpur University , Gorakhpur-273009, India
| |
Collapse
|
50
|
Dogan S, Unal M, Ozturk N, Yargicoglu P, Cort A, Spasojevic I, Batinic-Haberle I, Aslan M. Manganese porphyrin reduces retinal injury induced by ocular hypertension in rats. Exp Eye Res 2011; 93:387-96. [PMID: 21669199 PMCID: PMC3184467 DOI: 10.1016/j.exer.2011.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 05/12/2011] [Accepted: 05/17/2011] [Indexed: 10/18/2022]
Abstract
This study aimed to clarify the possible therapeutic benefit of preferential nitric oxide synthase (NOS) inhibition and catalytic antioxidant Mn (III) meso-tetrakis (N-n-hexylpyridinium-2-yl) porphyrin (MnTnHex-2-PyP(5+)) treatment in a rat model of elevated intraocular pressure (EIOP). Rats were randomly divided into different experimental groups which received either intraperitoneal MnTnHex-2-PyP(5+) (0.1 mg/kg/day), intragastric NOS inhibitor (S-methylthiourea: SMT; 5 mg/kg/day) or both agents for a period of 6 weeks. Ocular hypertension was induced by unilaterally cauterizing three episcleral vessels and the unoperated eye served as control. Neuroprotective effects of given treatments were determined via electrophysiological measurements of visual evoked potentials (VEP) while retina and vitreous levels of MnTnHex-2-PyP(5+) were measured via LC-MS/MS. Latencies of all VEP components (P(1), N(1), P(2), N(2), P(3)) were significantly prolonged (p < 0.05) in EIOP and returned to control levels following all three treatment protocols. Ocular hypertension significantly increased retinal protein nitration (p < 0.001) which returned to baseline levels in all treated groups. NOS-2 expression and nitrate/nitrite levels were significantly greater in non-treated rats with EIOP. Retinal TUNEL staining showed apoptosis in all ocular hypertensive rats. The presented data confirm the role of oxidative injury in EIOP and highlight the protective effect of MnTnHex-2-PyP(5+) treatment and NOS inhibition in ocular hypertension.
Collapse
Affiliation(s)
- Serdar Dogan
- Department of Biochemistry, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| | - Mustafa Unal
- Department of Ophthalmology, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| | - Nihal Ozturk
- Department of Biophysics, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| | - Piraye Yargicoglu
- Department of Biophysics, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| | - Aysegul Cort
- Department of Biochemistry, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| | - Ivan Spasojevic
- Department of Medicine Duke University Medical Center, Durham, NC. USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC. USA
| | - Mutay Aslan
- Department of Biochemistry, Akdeniz University Medical School, Campus, 07070, Antalya, Turkey
| |
Collapse
|