1
|
Abbas G, Alibrahim F, Kankouni R, Al-Belushi S, Al-Mutairi DA, Tovmasyan A, Batinic-Haberle I, Benov L. Effect of the nature of the chelated metal on the photodynamic activity of metalloporphyrins. Free Radic Res 2023; 57:487-499. [PMID: 38035627 DOI: 10.1080/10715762.2023.2288997] [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: 05/16/2023] [Accepted: 10/04/2023] [Indexed: 12/02/2023]
Abstract
Coordination of metal ions by the tetrapyrrolic macrocyclic ring of porphyrin-based photosensitizers (PSs) affects their photophysical properties and consequently, their photodynamic activity. Diamagnetic metals increase the singlet oxygen quantum yield while paramagnetic metals have the opposite effect. Since singlet oxygen is considered the main cell-damaging species in photodynamic therapy (PDT), the nature of the chelated cation would directly affect PDT efficacy. This expectation, however, is not always supported by experimental results and numerous exceptions have been reported. Understanding the effect of the chelated metal is hindered because different chelators were used. The aim of this work was to investigate the effect of the nature of chelated cation on the photophysical and photodynamic properties of metalloporphyrins, using the same tetrapyrrole core as a chelator of Ag(II), Cu(II), Fe(III), In(III), Mn(III), or Zn(II). Results demonstrated that with the exception of Ag(II), all paramagnetic metalloporphyrins were inefficient as generators of singlet oxygen and did not act as PSs. In contrast, the coordination of diamagnetic ions produced highly efficient PSs. The unexpected photodynamic activity of the Ag(II)-containing porphyrin was attributed to reduction of the chelated Ag(II) to Ag(I) or to demetallation of the complex, caused by cellular reductants and/or by exposure to light. Our results indicate that in biological systems, where PSs localize to various organelles and are subjected to the action of enzymes, reactive metabolites, and reducing or oxidizing agents, their physicochemical and photosensitizing properties change. Consequently, the photophysical properties alone cannot predict the anticancer efficacy of a PS.
Collapse
Affiliation(s)
- Ghadeer Abbas
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Fatemah Alibrahim
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Rawan Kankouni
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Sara Al-Belushi
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Dalal A Al-Mutairi
- Department of Pathology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Artak Tovmasyan
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
2
|
Inertness of Superoxide Dismutase Mimics Mn(II) Complexes Based on an Open-Chain Ligand, Bioactivity, and Detection in Intestinal Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3858122. [PMID: 35401918 PMCID: PMC8993562 DOI: 10.1155/2022/3858122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/03/2022] [Accepted: 01/29/2022] [Indexed: 12/22/2022]
Abstract
Oxidative stress is known to play a major role in the pathogenesis of inflammatory bowel diseases (IBDs), and, in particular, superoxide dismutase (SODs) defenses were shown to be weakened in patients suffering from IBDs. SOD mimics, also called SOD mimetics, as low-molecular-weight complexes reproducing the activity of SOD, constitute promising antioxidant catalytic metallodrugs in the context of IBDs. A Mn(II) complex SOD mimic (Mn1) based on an open-chain diaminoethane ligand exerting antioxidant and anti-inflammatory effects on an intestinal epithelial cellular model was shown to experience metal exchanges between the manganese center and metal ions present in the biological environment (such as Zn(II)) to some degrees. As the resulting complexes (mainly Zn(II)) were shown to be inactive, improving the kinetic inertness of Mn(II) complexes based on open-chain ligands is key to improve their bioactivity in a cellular context. We report here the study of three new Mn(II) complexes resulting from Mn1 functionalization with a cyclohexyl and/or a propyl group meant to limit, respectively, (a) metal exchanges and (b) deprotonation of an amine from the 1,2-diaminoethane central scaffold. The new manganese-based SOD mimics display a higher intrinsic SOD activity and also improved kinetic inertness in metal ion exchange processes (with Zn(II), Cu(II), Ni(II), and Co(II)). They were shown to provide anti-inflammatory and antioxidant effects in cells at lower doses than Mn1 (down to 10 μM). This improvement was due to their higher inertness against metal-assisted dissociation and not to different cellular overall accumulations. Based on its higher inertness, the SOD mimic containing both the propyl and the cyclohexyl moieties was suitable for intracellular detection and quantification by mass spectrometry, quantification, that was achieved by using a 13C-labeled Co-based analog of the SOD mimics as an external heavy standard.
Collapse
|
3
|
Maia CGC, de Araujo BCR, de Freitas-Marques MB, da Costa IF, Yoshida MI, da Nova Mussel W, Sebastião RDCO, Rebouças JS. Thermal Stability Kinetics and Shelf Life Estimation of the Redox-Active Therapeutic and Mimic of Superoxide Dismutase Enzyme, Mn(III) meso-Tetrakis( N-ethylpyridinium-2-yl)porphyrin Chloride (MnTE-2-PyPCl 5, BMX-010). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7003861. [PMID: 34912497 PMCID: PMC8668311 DOI: 10.1155/2021/7003861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin chloride (MnTE-2-PyPCl5, BMX-010, and AEOL10113) is among the most studied superoxide dismutase (SOD) mimics and redox-active therapeutics, being currently tested as a drug candidate in a phase II clinical trial on atopic dermatitis and itch. The thermal stability of active pharmaceutical ingredients (API) is useful for estimating the expiration date and shelf life of pharmaceutical products under various storage and handling conditions. The thermal decomposition and kinetic parameters of MnTE-2-PyPCl5 were determined by thermogravimetry (TG) under nonisothermal and isothermal conditions. The first thermal degradation pathway affecting Mn-porphyrin structural integrity and, thus, activity and bioavailability was associated with loss of ethyl chloride via N-dealkylation reaction. The thermal stability kinetics of the N-dealkylation process leading to MnTE-2-PyPCl5 decomposition was investigated by using isoconversional models and artificial neural network. The new multilayer perceptron (MLP) artificial neural network approach allowed the simultaneous study of ten solid-state kinetic models and showed that MnTE-2-PyPCl5 degradation is better explained by a combination of various mechanisms, with major contributions from the contraction models R1 and R2. The calculated activation energy values from isothermal and nonisothermal data were about 90 kJ mol-1 on average and agreed with one another. According to the R1 modelling of the isothermal decomposition data, the estimated shelf life value for 10% decomposition (t 90%) of MnTE-2-PyPCl5 at 25°C was approximately 17 years, which is consistent with the high solid-state stability of the compound. These results represent the first study on the solid-state decomposition kinetics of Mn(III) 2-N-alkylpyridylporphyrins, contributing to the development of this class of redox-active therapeutics and SOD mimics and providing supporting data to protocols on purification, handling, storage, formulation, expiration date, and general use of these compounds.
Collapse
Affiliation(s)
- Clarissa G. C. Maia
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Bárbara C. R. de Araujo
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31207-901, Brazil
| | - Maria B. de Freitas-Marques
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31207-901, Brazil
| | - Israel F. da Costa
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| | - Maria Irene Yoshida
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31207-901, Brazil
| | - Wagner da Nova Mussel
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31207-901, Brazil
| | - Rita de Cássia O. Sebastião
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31207-901, Brazil
| | - Júlio S. Rebouças
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa, PB 58051-900, Brazil
| |
Collapse
|
4
|
|
5
|
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
|
6
|
Faustova M, Nikolskaya E, Sokol M, Fomicheva M, Petrov R, Yabbarov N. Metalloporphyrins in Medicine: From History to Recent Trends. ACS APPLIED BIO MATERIALS 2020; 3:8146-8171. [PMID: 35019597 DOI: 10.1021/acsabm.0c00941] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The history of metalloporphyrins dates back more than 200 years ago. Metalloporphyrins are excellent catalysts, capable of forming supramolecular systems, participate in oxygen photosynthesis, transport, and used as contrast agents or superoxide dismutase mimetics. Today, metalloporphyrins represent complexes of conjugated π-electron system and metals from the entire periodic system. However, the effect of these compounds on living systems has not been fully understood, and researchers are exploring the properties of metalloporphyrins thereby extending their further application. This review provides an overview of the variety of metalloporphyrins that are currently used in different medicine fields and how metalloporphyrins became the subject of scientists' interest. Currently, metalloporphyrins utilization has expanded significantly, which gave us an opprotunuty to summarize recent progress in metalloporphyrins derivatives and prospects of their application in the treatment and diagnosis of different diseases.
Collapse
Affiliation(s)
- Mariia Faustova
- MIREA-Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454 Moscow, Russia.,N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena Nikolskaya
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia
| | - Maria Sokol
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| | - Margarita Fomicheva
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| | - Rem Petrov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia
| | - Nikita Yabbarov
- N. M. Emanuel Institute of Biochemical Physics of Russian Academy of Sciences, 119991 Moscow, Russia.,JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149 Moscow Russia
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Tovmasyan A, Bueno-Janice JC, Jaramillo MC, Sampaio RS, Reboucas JS, Kyui N, Benov L, Deng B, Huang TT, Tome ME, Spasojevic I, Batinic-Haberle I. Radiation-Mediated Tumor Growth Inhibition Is Significantly Enhanced with Redox-Active Compounds That Cycle with Ascorbate. Antioxid Redox Signal 2018; 29:1196-1214. [PMID: 29390861 PMCID: PMC6157436 DOI: 10.1089/ars.2017.7218] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/12/2018] [Accepted: 02/01/2018] [Indexed: 12/24/2022]
Abstract
AIMS We aim here to demonstrate that radiation (RT) enhances tumor sensitization by only those Mn complexes that are redox active and cycle with ascorbate (Asc), thereby producing H2O2 and utilizing it subsequently in protein S-glutathionylation in a glutathione peroxidase (GPx)-like manner. In turn, such compounds affect cellular redox environment, described by glutathione disulfide (GSSG)/glutathione (GSH) ratio, and tumor growth. To achieve our goal, we tested several Mn complexes of different chemical and physical properties in cellular and animal flank models of 4T1 breast cancer cell. Four other cancer cell lines were used to substantiate key findings. RESULTS Joint administration of cationic Mn porphyrin (MnP)-based redox active compounds, MnTE-2-PyP5+ or MnTnBuOE-2-PyP5+ with RT and Asc contributes to high H2O2 production in cancer cells and tumor, which along with high MnP accumulation in cancer cells and tumor induces the largest suppression of cell viability and tumor growth, while increasing GSSG/GSH ratio and levels of total S-glutathionylated proteins. Redox-inert MnP, MnTBAP3- and two other different types of redox-active Mn complexes (EUK-8 and M40403) were neither efficacious in the cellular nor in the animal model. Such outcome is in accordance with their inability to catalyze Asc oxidation and mimic GPx. INNOVATION We provided here the first evidence how structure-activity relationship between the catalytic potency and the redox properties of Mn complexes controls their ability to impact cellular redox environment and thus enhance the radiation and ascorbate-mediated tumor suppression. CONCLUSIONS The interplay between the accumulation of cationic MnPs and their potency as catalysts for oxidation of Asc, protein cysteines, and GSH controls the magnitude of their anticancer therapeutic effects.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | | | | | - Romulo S. Sampaio
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Julio S. Reboucas
- Departamento de Quimica, CCEN, Universidade Federal da Paraiba, Joao Pessoa, Brazil
| | - Natalia Kyui
- Canadian Economic Analysis Department, Bank of Canada, Ottawa, Canada
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait, Kuwait
| | - Brian Deng
- Palo Alto Veterans Institute for Research, Palo Alto, California
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California
- Geriatric Research. Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California
| | - Margaret E. Tome
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | - Ivan Spasojevic
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- PK/PD Core Laboratory, Pharmaceutical Research Shared Resource, Duke Cancer Institute, Durham, North Carolina
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
10
|
Conte-Daban A, Ambike V, Guillot R, Delsuc N, Policar C, Hureau C. A Metallo Pro-Drug to Target Cu II in the Context of Alzheimer's Disease. Chemistry 2018; 24:5095-5099. [PMID: 29334419 PMCID: PMC6120673 DOI: 10.1002/chem.201706049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 01/28/2023]
Abstract
Alzheimer's disease and oxidative stress are connected. In the present communication, we report the use of a MnII -based superoxide dismutase (SOD) mimic ([MnII (L)]+ , 1+ ) as a pro-drug candidate to target CuII -associated events, namely, CuII -induced formation of reactive oxygen species (ROS) and modulation of the amyloid-β (Aβ) peptide aggregation. Complex 1+ is able to remove CuII from Aβ, stop ROS and prevent alteration of Aβ aggregation as would do the corresponding free ligand LH. Using 1+ instead of LH in further biological applications would have the double advantage to avoid the cell toxicity of LH and to benefit from its proved SOD-like activity.
Collapse
Affiliation(s)
- Amandine Conte-Daban
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- Université de Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| | - Vinita Ambike
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR CNRS 8182, Bâtiments 420, Université Paris-Sud 11, Université Paris-Saclay, Rue du doyen Georges Poitou, 91405 Orsay cedex, France
| | - Régis Guillot
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, UMR CNRS 8182, Bâtiments 420, Université Paris-Sud 11, Université Paris-Saclay, Rue du doyen Georges Poitou, 91405 Orsay cedex, France
| | - Nicolas Delsuc
- Laboratoire des Biomolécules, Département de chimie, École normale supérieure, UPMC Univ. Paris 06, CNRS, PSL Research University, 24 rue Lhomond, 75005 Paris, France
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, CNRS, Laboratoire des Biomolécules (LBM), 75005 Paris, France
| | - Clotilde Policar
- Laboratoire des Biomolécules, Département de chimie, École normale supérieure, UPMC Univ. Paris 06, CNRS, PSL Research University, 24 rue Lhomond, 75005 Paris, France
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, CNRS, Laboratoire des Biomolécules (LBM), 75005 Paris, France
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077 Toulouse Cedex 4, France
- Université de Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France
| |
Collapse
|
11
|
Shin SW, Choi C, Lee GH, Son A, Kim SH, Park HC, Batinic-Haberle I, Park W. Mechanism of the Antitumor and Radiosensitizing Effects of a Manganese Porphyrin, MnHex-2-PyP. Antioxid Redox Signal 2017; 27:1067-1082. [PMID: 28358581 DOI: 10.1089/ars.2016.6889] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIMS Cationic manganese (Mn)-substituted N-pyridylporphyrin-based potent mimics of the family of superoxide dismutases (SODs) protect normal tissues from injury related to ionizing radiation (IR) by reducing levels of reactive oxygen and nitrogen species (ROS/RNS). Furthermore, Mn-porphyrins have demonstrated antitumor and radiosensitizing effects on cancer cells by promoting IR-induced tumor vasculature damage and apoptotic processes. In this study, we explored the underlying mechanisms of Mn-porphyrin-mediated tumor radiosensitization using murine mammary carcinoma 4T1 and melanoma B16 cells in vitro and in vivo. RESULTS Combination treatment with MnTnHex-2-PyP and IR substantially reduced cell viability, clonogenic cell survival, and DNA damage repair and synergistically increased IR-induced apoptosis of 4T1 and B16 cells. MnTnHex-2-PyP in combination with IR caused a significant delay in growth of 4T1 and B16 xenograft tumors. MnTnHex-2-PyP dose-dependently enhanced IR-mediated production of H2O2-derived species, but not superoxide. Catalase overexpression reversed MnTnHex-2-PyP-enhanced ROS production and apoptosis. Demonstrated suppression of phosphorylation of several mitogen-activated protein (MAP) kinases and activation of NF-κB by MnTnHex-2-PyP/IR, which presumably inhibited activation of the antiapoptotic pathway, are in agreement with our other data on the apoptosis of cancer cells. Innovation and Conclusions: MnTnHex-2-PyP exerted a radiosensitizing effect on 4T1 and B16 tumor models in vitro and in vivo via pro-oxidative actions and therefore bears a large therapeutic potential. When combined with IR, it attenuated DNA damage repair and triggered a shift from prosurvival pathways to apoptotic cell death, likely due to increased ROS production and disturbed cellular redox balance, acting at the level of nuclear factor κB (NF-κB). Antioxid. Redox Signal. 27, 1067-1082.
Collapse
Affiliation(s)
- Sung-Won Shin
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea.,2 Sungkyunkwan University School of Medicine , Seoul, Republic of Korea
| | - Changhoon Choi
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea
| | - Ga-Haeng Lee
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea
| | - Arang Son
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea
| | - Su-Hyeon Kim
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea
| | - Hee Chul Park
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea.,2 Sungkyunkwan University School of Medicine , Seoul, Republic of Korea
| | - Ines Batinic-Haberle
- 3 Department of Radiation Oncology, Duke University School of Medicine , Durham, North Carolina
| | - Won Park
- 1 Department of Radiation Oncology, Samsung Medical Center , Seoul, Republic of Korea.,2 Sungkyunkwan University School of Medicine , Seoul, Republic of Korea
| |
Collapse
|
12
|
Yin B, Barrionuevo G, Batinic-Haberle I, Sandberg M, Weber SG. Differences in Reperfusion-Induced Mitochondrial Oxidative Stress and Cell Death Between Hippocampal CA1 and CA3 Subfields Are Due to the Mitochondrial Thioredoxin System. Antioxid Redox Signal 2017; 27:534-549. [PMID: 28129719 PMCID: PMC5567420 DOI: 10.1089/ars.2016.6706] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS The susceptibility of CA1 over CA3 to damage from cerebral ischemia may be related to the differences in reactive oxygen species (ROS) production/removal between the two hippocampal subfields. We aimed to measure CA1/CA3 differences in net ROS production in real time in the first 30 min of reperfusion in pyramidal cells. We aimed to determine the underlying cause of the differential vulnerability of CA1 and CA3. RESULTS Real-time determinations of mitochondrial H2O2 and, independently, glutathione (GSH) redox status from roGFP-based probes in individual pyramidal cells in organotypic hippocampal cultures during oxygen-glucose deprivation (OGD)-reperfusion (RP) demonstrate a significantly more oxidizing environment during RP in CA1 than CA3 mitochondria. Protein levels (immunohistochemistry and Western blots), roGFP2-based probe measurements during controlled mitochondrial production of ROS, and thioredoxin reductase (TrxR) inhibition by auranofin are consistent with a more effective mitochondrial thioredoxin (Trx) system in CA3. Inhibition of TrxR eliminates the differences in redox status and cell death between the regions. Overexpression of cytosolic Trx1 does not influence mitochondrial H2O2 production. INNOVATION Real-time changes of mitochondrial H2O2 and GSH in tissue cultures during early RP, and also during controlled production of superoxide and peroxide, reveal significant differences between CA1 and CA3. The mitochondrial Trx system is responsible for the observed differences during RP as well as for delayed cell death 18 h afterward. CONCLUSION Greater mitochondrial Trx efficacy in CA3 pyramidal cells results in less vulnerability to ischemia/reperfusion because of the less oxidizing environment in CA3 mitochondria during RP. Antioxid. Redox Signal. 27, 534-549.
Collapse
Affiliation(s)
- Bocheng Yin
- 1 Department of Chemistry, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Germán Barrionuevo
- 2 Department of Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Ines Batinic-Haberle
- 3 Department of Radiation Oncology, Duke University Medical Center , North Carolina
| | - Mats Sandberg
- 4 Department of Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg , Göteborg, Sweden
| | - Stephen G Weber
- 1 Department of Chemistry, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|