1
|
Di Y, Deng R, Liu Z, Mao Y, Gao Y, Zhao Q, Wang S. Optimized strategies of ROS-based nanodynamic therapies for tumor theranostics. Biomaterials 2023; 303:122391. [PMID: 37995457 DOI: 10.1016/j.biomaterials.2023.122391] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/25/2023]
Abstract
Reactive oxygen species (ROS) play a crucial role in regulating the metabolism of tumor growth, metastasis, death and other biological processes. ROS-based nanodynamic therapies (NDTs) are becoming attractive due to non-invasive, low side effects and tumor-specific advantages. NDTs have rapidly developed into numerous branches, such as photodynamic therapy, chemodynamic therapy, sonodynamic therapy and so on. However, the complexity of the tumor microenvironment and the limitations of existing sensitizers have greatly restricted the therapeutic effects of NDTs, which heavily rely on ROS levels. To address the limitations of NDTs, various strategies have been developed to increase ROS yield, which is an urgent aspect for the positive development of NDTs. In this review, the nanodynamic potentiation strategies in terms of unique properties and universalities of NDTs are comprehensively outlined. We mainly summarize the current dilemmas faced by each NDT and the respective solutions. Meanwhile, the NDTs universalities-based potentiation strategies and NDTs-based combined treatments are elaborated. Finally, we conclude with a discussion of the key issues and challenges faced in the development and clinical transformation of NDTs.
Collapse
Affiliation(s)
- Yifan Di
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Ruizhu Deng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Zhu Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Yikun Gao
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| |
Collapse
|
2
|
Russell TM, Richardson DR. The good Samaritan glutathione-S-transferase P1: An evolving relationship in nitric oxide metabolism mediated by the direct interactions between multiple effector molecules. Redox Biol 2023; 59:102568. [PMID: 36563536 PMCID: PMC9800640 DOI: 10.1016/j.redox.2022.102568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Glutathione-S-transferases (GSTs) are phase II detoxification isozymes that conjugate glutathione (GSH) to xenobiotics and also suppress redox stress. It was suggested that GSTs have evolved not to enhance their GSH affinity, but to better interact with and metabolize cytotoxic nitric oxide (NO). The interactions between NO and GSTs involve their ability to bind and store NO as dinitrosyl-dithiol iron complexes (DNICs) within cells. Additionally, the association of GSTP1 with inducible nitric oxide synthase (iNOS) results in its inhibition. The function of NO in vasodilation together with studies associating GSTM1 or GSTT1 null genotypes with preeclampsia, additionally suggests an intriguing connection between NO and GSTs. Furthermore, suppression of c-Jun N-terminal kinase (JNK) activity occurs upon increased levels of GSTP1 or NO that decreases transcription of JNK target genes such as c-Jun and c-Fos, which inhibit apoptosis. This latter effect is mediated by the direct association of GSTs with MAPK proteins. GSTP1 can also inhibit nuclear factor kappa B (NF-κB) signaling through its interactions with IKKβ and Iκα, resulting in decreased iNOS expression and the stimulation of apoptosis. It can be suggested that the inhibitory activity of GSTP1 within the JNK and NF-κB pathways may be involved in crosstalk between survival and apoptosis pathways and modulating NO-mediated ROS generation. These studies highlight an innovative role of GSTs in NO metabolism through their interaction with multiple effector proteins, with GSTP1 functioning as a "good Samaritan" within each pathway to promote favorable cellular conditions and NO levels.
Collapse
Affiliation(s)
- Tiffany M Russell
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Australia.
| |
Collapse
|
3
|
Bouabid C, Rabhi S, Thedinga K, Barel G, Tnani H, Rabhi I, Benkahla A, Herwig R, Guizani-Tabbane L. Host M-CSF induced gene expression drives changes in susceptible and resistant mice-derived BMdMs upon Leishmania major infection. Front Immunol 2023; 14:1111072. [PMID: 37187743 PMCID: PMC10175952 DOI: 10.3389/fimmu.2023.1111072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Leishmaniases are a group of diseases with different clinical manifestations. Macrophage-Leishmania interactions are central to the course of the infection. The outcome of the disease depends not only on the pathogenicity and virulence of the parasite, but also on the activation state, the genetic background, and the underlying complex interaction networks operative in the host macrophages. Mouse models, with mice strains having contrasting behavior in response to parasite infection, have been very helpful in exploring the mechanisms underlying differences in disease progression. We here analyzed previously generated dynamic transcriptome data obtained from Leishmania major (L. major) infected bone marrow derived macrophages (BMdMs) from resistant and susceptible mouse. We first identified differentially expressed genes (DEGs) between the M-CSF differentiated macrophages derived from the two hosts, and found a differential basal transcriptome profile independent of Leishmania infection. These host signatures, in which 75% of the genes are directly or indirectly related to the immune system, may account for the differences in the immune response to infection between the two strains. To gain further insights into the underlying biological processes induced by L. major infection driven by the M-CSF DEGs, we mapped the time-resolved expression profiles onto a large protein-protein interaction (PPI) network and performed network propagation to identify modules of interacting proteins that agglomerate infection response signals for each strain. This analysis revealed profound differences in the resulting responses networks related to immune signaling and metabolism that were validated by qRT-PCR time series experiments leading to plausible and provable hypotheses for the differences in disease pathophysiology. In summary, we demonstrate that the host's gene expression background determines to a large degree its response to L. major infection, and that the gene expression analysis combined with network propagation is an effective approach to help identifying dynamically altered mouse strain-specific networks that hold mechanistic information about these contrasting responses to infection.
Collapse
Affiliation(s)
- Cyrine Bouabid
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- Faculty of Sciences of Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Sameh Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Kristina Thedinga
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Gal Barel
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Hedia Tnani
- Laboratory de BioInformatic, BioMathematic and BioStatistic (BIMS), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Imen Rabhi
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- Higher Institute of Biotechnology at Sidi-Thabet (ISBST), Biotechnopole Sidi-Thabet- University of Manouba, Sidi-Thabet, Tunisia
| | - Alia Benkahla
- Laboratory de BioInformatic, BioMathematic and BioStatistic (BIMS), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Ralf Herwig
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lamia Guizani-Tabbane
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules (PMBB), Institut Pasteur de Tunis, Tunis, Tunisia
- *Correspondence: Lamia Guizani-Tabbane,
| |
Collapse
|
4
|
Vanin AF. Positive (Regulatory) and Negative (Cytotoxic) Effects of Dinitrosyl Iron Complexes on Living Organisms. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1367-1386. [PMID: 36509730 PMCID: PMC9672603 DOI: 10.1134/s0006297922110153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022]
Abstract
The proposed in our studies mechanism of dinitrosyl iron complex (DNIC) formation through the main step of disproportionation of two NO molecules in complex with Fe2+ ion leads to emergence of the resonance structure of dinitrosyl-iron fragment of DNIC, [Fe2+(NO)(NO+)]. The latter allowed suggesting capacity of these complexes to function as donor of both neutral NO molecules as well as nitrosonium cations (NO+), which has been demonstrated in experiments. Analysis of biological activity of DNICs with thiol-containing ligands presented in this review demonstrates that NO molecules and nitrosonium cations released from the complexes exert respectively positive (regulatory) and negative (cytotoxic) effects on living organisms. It has been suggested to use dithiocarbamate derivatives to enhance selective release of nitrosonium cations from DNIC in living organisms followed by simultaneous incorporation of the released NO molecules into the biologically non-active mononitrosyl iron complexes with dithiocarbamate derivatives.
Collapse
Affiliation(s)
- Anatoly F Vanin
- Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
5
|
Russell TM, Richardson DR. Glutathione-S-Transferases as Potential Targets for Modulation of Nitric Oxide-Mediated Vasodilation. Biomolecules 2022; 12:biom12091292. [PMID: 36139130 PMCID: PMC9496536 DOI: 10.3390/biom12091292] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Glutathione-S-transferases (GSTs) are highly promiscuous in terms of their interactions with multiple proteins, leading to various functions. In addition to their classical detoxification roles with multi-drug resistance-related protein-1 (MRP1), more recent studies have indicated the role of GSTs in cellular nitric oxide (NO) metabolism. Vasodilation is classically induced by NO through its interaction with soluble guanylate cyclase. The ability of GSTs to biotransform organic nitrates such as nitroglycerin for NO generation can markedly modulate vasodilation, with this effect being prevented by specific GST inhibitors. Recently, other structurally distinct pro-drugs that generate NO via GST-mediated catalysis have been developed as anti-cancer agents and also indicate the potential of GSTs as suitable targets for pharmaceutical development. Further studies investigating GST biochemistry could enhance our understanding of NO metabolism and lead to the generation of novel and innovative vasodilators for clinical use.
Collapse
Affiliation(s)
- Tiffany M. Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Brisbane 4111, Australia
| | - Des R. Richardson
- Department of Pathology and Biological Responses, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
- Correspondence: ; Tel.: +61-7-3735-7549
| |
Collapse
|
6
|
Abedi M, Rahgozar S. Puzzling Out Iron Complications in Cancer Drug Resistance. Crit Rev Oncol Hematol 2022; 178:103772. [PMID: 35914667 DOI: 10.1016/j.critrevonc.2022.103772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/09/2022] Open
Abstract
Iron metabolism are frequently disrupted in cancer. Patients with cancer are prone to anemia and receive transfusions frequently; the condition which results in iron overload, contributing to serious therapeutic complications. Iron is introduced as a carcinogen that may increase tumor growth. However, investigations regarding its impact on response to chemotherapy, particularly the induction of drug resistance are still limited. Here, iron contribution to cell signaling and various molecular mechanisms underlying iron-mediated drug resistance are described. A dual role of this vital element in cancer treatment is also addressed. On one hand, the need to administer iron chelators to surmount iron overload and improve the sensitivity of tumor cells to chemotherapy is discussed. On the other hand, the necessary application of iron as a therapeutic option by iron-oxide nanoparticles or ferroptosis inducers is explained. Authors hope that this paper can help unravel the clinical complications related to iron in cancer therapy.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
7
|
Li F, Wang H, Chen H, Guo J, Dang X, Ru Y, Wang H. Mechanism of Ferroptosis and Its Role in Spinal Cord Injury. Front Neurol 2022; 13:926780. [PMID: 35756929 PMCID: PMC9218271 DOI: 10.3389/fneur.2022.926780] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a non-necrotic form of regulated cell death (RCD) that is primarily characterized by iron-dependent membrane lipid peroxidation and is regulated by cysteine transport, glutathione synthesis, and glutathione peroxidase 4 function as well as other proteins including ferroptosis suppressor protein 1. It has been found that ferroptosis played an important role in many diseases, such as neurodegenerative diseases and ischemia-reperfusion injury. Spinal cord injury (SCI), especially traumatic SCI, is an urgent problem worldwide due to its high morbidity and mortality, as well as the destruction of functions of the human body. Various RCDs, including ferroptosis, are found in SCI. Different from necrosis, since RCD is a form of cell death regulated by various molecular mechanisms in cells, the study of the role played by RCD in SCI will contribute to a deeper understanding of the pathophysiological process, as well as the treatment and functional recovery. The present review mainly introduces the main mechanism of ferroptosis and its role in SCI, so as to provide a new idea for further exploration.
Collapse
Affiliation(s)
- Fei Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haifan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Chen
- Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Jianing Guo
- Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Xiaoqian Dang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Ru
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Haoyu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Liu T, Schroeder H, Power GG, Blood AB. A physiologically relevant role for NO stored in vascular smooth muscle cells: A novel theory of vascular NO signaling. Redox Biol 2022; 53:102327. [PMID: 35605454 PMCID: PMC9126848 DOI: 10.1016/j.redox.2022.102327] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/16/2022] [Accepted: 04/29/2022] [Indexed: 01/16/2023] Open
Abstract
S-nitrosothiols (SNO), dinitrosyl iron complexes (DNIC), and nitroglycerine (NTG) dilate vessels via activation of soluble guanylyl cyclase (sGC) in vascular smooth muscle cells. Although these compounds are often considered to be nitric oxide (NO) donors, attempts to ascribe their vasodilatory activity to NO-donating properties have failed. Even more puzzling, many of these compounds have vasodilatory potency comparable to or even greater than that of NO itself, despite low membrane permeability. This raises the question: How do these NO adducts activate cytosolic sGC when their NO moiety is still outside the cell? In this review, we classify these compounds as ‘nitrodilators’, defined by their potent NO-mimetic vasoactivities despite not releasing requisite amounts of free NO. We propose that nitrodilators activate sGC via a preformed nitrodilator-activated NO store (NANOS) found within the vascular smooth muscle cell. We reinterpret vascular NO handling in the framework of this NANOS paradigm, and describe the knowledge gaps and perspectives of this novel model.
Collapse
|
9
|
Singh N, Bhatla SC. Heme oxygenase-nitric oxide crosstalk-mediated iron homeostasis in plants under oxidative stress. Free Radic Biol Med 2022; 182:192-205. [PMID: 35247570 DOI: 10.1016/j.freeradbiomed.2022.02.034] [Citation(s) in RCA: 3] [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: 11/29/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 12/22/2022]
Abstract
Plant growth under abiotic stress conditions significantly enhances intracellular generation of reactive oxygen species (ROS). Oxidative status of plant cells is directly affected by the modulation of iron homeostasis. Among mammals and plants, heme oxygenase-1 (HO-1) is a well-known antioxidant enzyme. It catalyzes oxygenation of heme, thereby producing Fe2+, CO and biliverdin as byproducts. The antioxidant potential of HO-1 is primarily due to its catalytic reaction byproducts. Biliverdin and bilirubin possess conjugated π-electrons which escalate the ability of these biomolecules to scavenge free radicals. CO also enhances the ROS scavenging ability of plants cells by upregulating catalase and peroxidase activity. Enhanced expression of HO-1 in plants under oxidative stress accompanies sequestration of iron in specialized iron storage proteins localized in plastids and mitochondria, namely ferritin for Fe3+ storage and frataxin for storage of Fe-S clusters, respectively. Nitric oxide (NO) crosstalks with HO-1 at multiple levels, more so in plants under oxidative stress, in order to maintain intracellular iron status. Formation of dinitrosyl-iron complexes (DNICs) significantly prevents Fenton reaction during oxidative stress. DNICs also release NO upon dissociation in target cells over long distance in plants. They also function as antioxidants against superoxide anions and lipidic free radicals. A number of NO-modulated transcription factors also facilitate iron homeostasis in plant cells. Plants facing oxidative stress exhibit modulation of lateral root formation by HO-1 through NO and auxin-dependent pathways. The present review provides an in-depth analysis of the structure-function relationship of HO-1 in plants and mammals, correlating them with their adaptive mechanisms of survival under stress.
Collapse
Affiliation(s)
- Neha Singh
- Department of Botany, Gargi College, University of Delhi, India.
| | - Satish C Bhatla
- Laboratory of Plant Physiology and Biochemistry, Department of Botany, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
10
|
Lehnert N, Kim E, Dong HT, Harland JB, Hunt AP, Manickas EC, Oakley KM, Pham J, Reed GC, Alfaro VS. The Biologically Relevant Coordination Chemistry of Iron and Nitric Oxide: Electronic Structure and Reactivity. Chem Rev 2021; 121:14682-14905. [PMID: 34902255 DOI: 10.1021/acs.chemrev.1c00253] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) is an important signaling molecule that is involved in a wide range of physiological and pathological events in biology. Metal coordination chemistry, especially with iron, is at the heart of many biological transformations involving NO. A series of heme proteins, nitric oxide synthases (NOS), soluble guanylate cyclase (sGC), and nitrophorins, are responsible for the biosynthesis, sensing, and transport of NO. Alternatively, NO can be generated from nitrite by heme- and copper-containing nitrite reductases (NIRs). The NO-bearing small molecules such as nitrosothiols and dinitrosyl iron complexes (DNICs) can serve as an alternative vehicle for NO storage and transport. Once NO is formed, the rich reaction chemistry of NO leads to a wide variety of biological activities including reduction of NO by heme or non-heme iron-containing NO reductases and protein post-translational modifications by DNICs. Much of our understanding of the reactivity of metal sites in biology with NO and the mechanisms of these transformations has come from the elucidation of the geometric and electronic structures and chemical reactivity of synthetic model systems, in synergy with biochemical and biophysical studies on the relevant proteins themselves. This review focuses on recent advancements from studies on proteins and model complexes that not only have improved our understanding of the biological roles of NO but also have provided foundations for biomedical research and for bio-inspired catalyst design in energy science.
Collapse
Affiliation(s)
- Nicolai Lehnert
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Eunsuk Kim
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Hai T Dong
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jill B Harland
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Andrew P Hunt
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Elizabeth C Manickas
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Kady M Oakley
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - John Pham
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Garrett C Reed
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Victor Sosa Alfaro
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| |
Collapse
|
11
|
Wu WY, Tsai ML, Lai YA, Hsieh CH, Liaw WF. NO Reduction to N 2O Triggered by a Dinuclear Dinitrosyl Iron Complex via the Associated Pathways of Hyponitrite Formation and NO Disproportionation. Inorg Chem 2021; 60:15874-15889. [PMID: 34015211 DOI: 10.1021/acs.inorgchem.1c00541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In spite of the comprehensive study of the metal-mediated conversion of NO to N2O disclosing the conceivable processes/mechanism in biological and biomimetic studies, in this study, the synthesis cycles and mechanism of NO reduction to N2O triggered by the electronically localized dinuclear {Fe(NO)2}10-{Fe(NO)2}9 dinitrosyl iron complex (DNIC) [Fe(NO)2(μ-bdmap)Fe(NO)2(THF)] (1) (bdmap = 1,3- bis(dimethylamino)-2-propanolate) were investigated in detail. Reductive conversion of NO to N2O triggered by complex 1 in the presence of exogenous ·NO occurs via the simultaneous formation of hyponitrite-bound {[Fe2(NO)4(μ-bdmap)]2(κ4-N2O2)} (2) and [NO2]--bridged [Fe2(NO)4(μ-bdmap)(μ-NO2)] (3) (NO disproportionation yielding N2O and complex 3). EPR/IR spectra, single-crystal X-ray diffraction, and the electrochemical study uncover the reversible redox transformation of {Fe(NO)2}9-{Fe(NO)2}9 [Fe2(NO)4(μ-bdmap)(μ-OC4H8)]+ (7) ↔ {Fe(NO)2}10-{Fe(NO)2}9 1 ↔ {Fe(NO)2}10-{Fe(NO)2}10 [Fe(NO)2(μ-bdmap)Fe(NO)2]- (6) and characterize the formation of complex 1. Also, the synthesis study and DFT computation feature the detailed mechanism of electronically localized {Fe(NO)2}10-{Fe(NO)2}9 DNIC 1 reducing NO to N2O via the associated hyponitrite-formation and NO-disproportionation pathways. Presumably, the THF-bound {Fe(NO)2}9 unit of electronically localized {Fe(NO)2}10-{Fe(NO)2}9 complex 1 served as an electron buffering reservoir for accommodating electron redistribution, and the {Fe(NO)2}10 unit of complex 1 acted as an electron-transfer channel to drive exogeneous ·NO coordination to yield proposed relay intermediate κ2-N,O-[NO]--bridged [Fe2(NO)4(μ-bdmap)(μ-NO)] (A) for NO reduction to N2O.
Collapse
Affiliation(s)
- Wun-Yan Wu
- Department of Chemistry and Frontier Research Center of Fundamental and Applied Science of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ming-Li Tsai
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Yi-An Lai
- Department of Chemistry and Frontier Research Center of Fundamental and Applied Science of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chieh-Hsin Hsieh
- Department of Chemistry and Frontier Research Center of Fundamental and Applied Science of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry and Frontier Research Center of Fundamental and Applied Science of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
12
|
Truzzi DR, Medeiros NM, Augusto O, Ford PC. Dinitrosyl Iron Complexes (DNICs). From Spontaneous Assembly to Biological Roles. Inorg Chem 2021; 60:15835-15845. [PMID: 34014639 DOI: 10.1021/acs.inorgchem.1c00823] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dinitrosyl iron complexes (DNICs) are spontaneously and rapidly generated in cells. Their assembly requires nitric oxide (NO), biothiols, and nonheme iron, either labile iron or iron-sulfur clusters. Despite ubiquitous detection by electron paramagnetic resonance in NO-producing cells, the DNIC's chemical biology remains only partially understood. In this Forum Article, we address the reaction mechanisms for endogenous DNIC formation, with a focus on a labile iron pool as the iron source. The capability of DNICs to promote S-nitrosation is discussed in terms of S-nitrosothiol generation associated with the formation and chemical reactivity of DNICs. We also highlight how elucidation of the chemical reactivity and the dynamics of DNICs combined with the development of detection/quantification methods can provide further information regarding their participation in physiological and pathological processes.
Collapse
Affiliation(s)
- Daniela R Truzzi
- Departamento de Bioquímica, Instituto de Química de São Paulo, Universidade de São Paulo, Caixa Postal 26077, CEP05513-970 São Paulo, São Paulo, Brazil
| | - Nathalia M Medeiros
- Departamento de Bioquímica, Instituto de Química de São Paulo, Universidade de São Paulo, Caixa Postal 26077, CEP05513-970 São Paulo, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica, Instituto de Química de São Paulo, Universidade de São Paulo, Caixa Postal 26077, CEP05513-970 São Paulo, São Paulo, Brazil
| | - Peter C Ford
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California 93106-9510, United States
| |
Collapse
|
13
|
The Relationship of Glutathione- S-Transferase and Multi-Drug Resistance-Related Protein 1 in Nitric Oxide (NO) Transport and Storage. Molecules 2021; 26:molecules26195784. [PMID: 34641326 PMCID: PMC8510172 DOI: 10.3390/molecules26195784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
Nitric oxide is a diatomic gas that has traditionally been viewed, particularly in the context of chemical fields, as a toxic, pungent gas that is the product of ammonia oxidation. However, nitric oxide has been associated with many biological roles including cell signaling, macrophage cytotoxicity, and vasodilation. More recently, a model for nitric oxide trafficking has been proposed where nitric oxide is regulated in the form of dinitrosyl-dithiol-iron-complexes, which are much less toxic and have a significantly greater half-life than free nitric oxide. Our laboratory has previously examined this hypothesis in tumor cells and has demonstrated that dinitrosyl-dithiol-iron-complexes are transported and stored by multi-drug resistance-related protein 1 and glutathione-S-transferase P1. A crystal structure of a dinitrosyl-dithiol-iron complex with glutathione-S-transferase P1 has been solved that demonstrates that a tyrosine residue in glutathione-S-transferase P1 is responsible for binding dinitrosyl-dithiol-iron-complexes. Considering the roles of nitric oxide in vasodilation and many other processes, a physiological model of nitric oxide transport and storage would be valuable in understanding nitric oxide physiology and pathophysiology.
Collapse
|
14
|
Chen YC, Chen YH, Chiu H, Ko YH, Wang RT, Wang WP, Chuang YJ, Huang CC, Lu TT. Cell-Penetrating Delivery of Nitric Oxide by Biocompatible Dinitrosyl Iron Complex and Its Dermato-Physiological Implications. Int J Mol Sci 2021; 22:ijms221810101. [PMID: 34576264 PMCID: PMC8469893 DOI: 10.3390/ijms221810101] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
After the discovery of endogenous dinitrosyl iron complexes (DNICs) as a potential biological equivalent of nitric oxide (NO), bioinorganic engineering of [Fe(NO)2] unit has emerged to develop biomimetic DNICs [(NO)2Fe(L)2] as a chemical biology tool for controlled delivery of NO. For example, water-soluble DNIC [Fe2(μ-SCH2CH2OH)2(NO)4] (DNIC-1) was explored for oral delivery of NO to the brain and for the activation of hippocampal neurogenesis. However, the kinetics and mechanism for cellular uptake and intracellular release of NO, as well as the biocompatibility of synthetic DNICs, remain elusive. Prompted by the potential application of NO to dermato-physiological regulations, in this study, cellular uptake and intracellular delivery of DNIC [Fe2(μ-SCH2CH2COOH)2(NO)4] (DNIC-2) and its regulatory effect/biocompatibility toward epidermal cells were investigated. Upon the treatment of DNIC-2 to human fibroblast cells, cellular uptake of DNIC-2 followed by transformation into protein-bound DNICs occur to trigger the intracellular release of NO with a half-life of 1.8 ± 0.2 h. As opposed to the burst release of extracellular NO from diethylamine NONOate (DEANO), the cell-penetrating nature of DNIC-2 rationalizes its overwhelming efficacy for intracellular delivery of NO. Moreover, NO-delivery DNIC-2 can regulate cell proliferation, accelerate wound healing, and enhance the deposition of collagen in human fibroblast cells. Based on the in vitro and in vivo biocompatibility evaluation, biocompatible DNIC-2 holds the potential to be a novel active ingredient for skincare products.
Collapse
Affiliation(s)
- Yu-Chieh Chen
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
| | - Han Chiu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
| | - Yi-Hsuan Ko
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Ruei-Ting Wang
- CHLITINA Research and Development Center, CHLITINA Holding Ltd., Taipei 10073, Taiwan; (R.-T.W.); (W.-P.W.)
| | - Wei-Ping Wang
- CHLITINA Research and Development Center, CHLITINA Holding Ltd., Taipei 10073, Taiwan; (R.-T.W.); (W.-P.W.)
| | - Yung-Jen Chuang
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
- Correspondence: (C.-C.H.); (T.-T.L.)
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
- Correspondence: (C.-C.H.); (T.-T.L.)
| |
Collapse
|
15
|
Rational Design of Biomolecules/Polymer Hybrids by Reversible Deactivation Radical Polymerization (RDRP) for Biomedical Applications. CHINESE JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1007/s10118-021-2543-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
16
|
Antioxidant Effect of Hydroxytyrosol, Hydroxytyrosol Acetate and Nitrohydroxytyrosol in a Rat MPP + Model of Parkinson's Disease. Neurochem Res 2021; 46:2923-2935. [PMID: 34260002 DOI: 10.1007/s11064-021-03379-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/03/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
3,4-Dihydroxyphenyl ethanol, known as hydroxytyrosol (HTy), is a phenylpropanoid found in diverse vegetable species. Several studies have demonstrated that HTy is a potent antioxidant. Thus, our study is aimed to evaluate the antioxidant effect of HTy and its derivatives, hydroxytyrosol acetate (HTyA) and nitrohydroxytyrosol (HTyN), in a model of oxidative stress induced by 1-methyl-4-phenylpyridinium (MPP+) in rats. Rats were administered intravenously (i.v.) in the tail with 1 mL saline solution or polyphenol compound (1.5 mg/kg) 5 min before intrastriatal infusion of 10 µg MPP+/8 µL. We found that rats injured with MPP+, pretreatment with HTy, HTyA or HTyN significantly decreased ipsilateral turns. This result was consistent with a significant preservation of striatal dopamine levels and decreased lipid fluorescence products (LFP), a marker of oxidative stress. Brain GSH/GSSG ratio, from rats pretreated with HTy or HTyN showed a significant preservation of that marker, decreased as a consequence of MPP+-induced oxidative damage. These results show an antioxidant effect of HTy, HTyA and HTyN in the MPP+ model of Parkinson's disease in the rat.
Collapse
|
17
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
18
|
Vanin AF. How is Nitric Oxide (NO) Converted into Nitrosonium Cations (NO +) in Living Organisms? (Based on the Results of Optical and EPR Analyses of Dinitrosyl Iron Complexes with Thiol-Containing Ligands). APPLIED MAGNETIC RESONANCE 2020; 51:851-876. [PMID: 33100585 PMCID: PMC7572240 DOI: 10.1007/s00723-020-01270-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The present work provides theoretical and experimental foundations for the ability of dinitrosyl iron complexes (DNICs) with thiol-containing ligands to be not only the donors of neutral NO molecules, but also the donors of nitrosonium cations (NO+) in living organisms ensuring S-nitrosation of various proteins and low-molecular-weight compounds. It is proposed that the emergence of those cations in DNICs is related to disproportionation reaction of NO molecules, initiated by their binding with Fe2+ ions (two NO molecules per one ion). At the same time, possible hydrolysis of iron-bound nitrosonium cations is prevented by the electron density transition to nitrosonium cations from sulfur atoms of thiol-containing ligands, which are included in the coordination sphere of iron. It allows supposing that iron in iron-nitrosyl complexes of DNICs has a d 7 electronic configuration. This supposition is underpinned by experimental data revealing that a half of nitrosyl ligands are converted into S-nitrosothiols (RSNOs) when those complexes decompose, with the other half of those ligands released in the form of neutral NO molecules.
Collapse
Affiliation(s)
- Anatoly F. Vanin
- Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| |
Collapse
|
19
|
Wang M, Gong J, Bhullar NK. Iron deficiency triggered transcriptome changes in bread wheat. Comput Struct Biotechnol J 2020; 18:2709-2722. [PMID: 33101609 PMCID: PMC7550799 DOI: 10.1016/j.csbj.2020.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022] Open
Abstract
A series of complex transport, storage and regulation mechanisms control iron metabolism and thereby maintain iron homeostasis in plants. Despite several studies on iron deficiency responses in different plant species, these mechanisms remain unclear in the allohexaploid wheat, which is the most widely cultivated commercial crop. We used RNA sequencing to reveal transcriptomic changes in the wheat flag leaves and roots, when subjected to iron limited conditions. We identified 5969 and 2591 differentially expressed genes (DEGs) in the flag leaves and roots, respectively. Genes involved in the synthesis of iron ligands i.e., nicotianamine (NA) and deoxymugineic acid (DMA) were significantly up-regulated during iron deficiency. In total, 337 and 635 genes encoding transporters exhibited altered expression in roots and flag leaves, respectively. Several genes related to MAJOR FACILITATOR SUPERFAMILY (MFS), ATP-BINDING CASSETTE (ABC) transporter superfamily, NATURAL RESISTANCE ASSOCIATED MACROPHAGE PROTEIN (NRAMP) family and OLIGOPEPTIDE TRANSPORTER (OPT) family were regulated, indicating their important roles in combating iron deficiency stress. Among the regulatory factors, the genes encoding for transcription factors of BASIC HELIX-LOOP-HELIX (bHLH) family were highly up-regulated in both roots and the flag leaves. The jasmonate biosynthesis pathway was significantly altered but with notable expression differences between roots and flag leaves. Homoeologs expression and induction bias analysis revealed subgenome specific differential expression. Our findings provide an integrated overview on regulated molecular processes in response to iron deficiency stress in wheat. This information could potentially serve as a guideline for breeding iron deficiency stress tolerant crops as well as for designing appropriate wheat iron biofortification strategies.
Collapse
Key Words
- 3-HMA, 3-hydroxymugineic acid
- ABC, ATP-BINDING CASSETTE
- ACC, 1-aminocyclopropane-1-carboxylate
- AEC, AUXIN EFFLUX CARRIER
- AOC, ALLENE OXIDE CYCLASE
- AOS, ALLENE OXIDE SYNTHASE
- AQP, AQUAPORIN
- AVA, avenic acid
- DEGs, differentially expressed genes
- DMA, deoxymugineic acid
- DMAS, DEOXYMUGINEIC ACID SYNTHASE
- DPA, days post anthesis
- ERF, ETHYLENE-RESPONSIVE FACTOR
- FAD, FATTY ACID DESATURASE
- FDR, false discovery rate
- FIT, FER-LIKE IRON DEFICIENCY-INDUCED TRANSCRIPTION FACTOR
- FRO, FERRIC REDUCTASE OXIDASE
- GCN, gene co-expression network
- GO, Gene ontology
- GSH, GLUTATHIONE
- HC, high confidence
- HMA, HEAVY METAL-ASSOCIATED
- IDE, iron deficiency-responsive cis-acting element
- IDEF, IDE BINDING FACTOR
- IHW, independent hypothesis weighting
- ILR3, IAA‐LEUCINE RESISTANT3
- IREG/FPN, IRON REGULATED PROTEIN/FERROPORTIN
- IRT1, IRON-REGULATED TRANSPORTER
- Iron deficiency
- Iron, Fe
- JAs, jasmonates
- JMT, JASMONATE O-METHYLTRANSFERASE
- KAT, 3-KETOACYL-COA THIOLASE
- LOX, LIPOXYGENASE
- MA, mugineic acid
- MATE, MULTI ANTIMICROBIAL EXTRUSION PROTEIN
- MFS, MAJOR FACILITATOR SUPERFAMILY
- MRP, MULTIDRUG RESISTANCE PROTEIN
- MT, METALLOTHIONEIN
- NA, nicotianamine
- NAAT, NICOTIANAMINE AMINOTRANSFERASE
- NAC, NO APICAL MERISTEM (NAM)/ARABIDOPSIS TRANSCRIPTION ACTIVATION FACTOR (ATAF)/CUP-SHAPED COTYLEDON (CUC)
- NAS, NICOTIANAMINE SYNTHASE
- NRAMP, NATURAL RESISTANCE ASSOCIATED MACROPHAGE PROTEIN
- NRT1/PTR, NITRATE TRANSPORTER 1/PEPTIDE TRANSPORTER
- OPCL, 4-COUMARATE COA LIGASE
- OPR, 12-OXOPHYTODIENOATE REDUCTASE
- OPT, OLIGOPEPTIDE TRANSPORTER
- PDR, PLEIOTROPIC DRUG RESISTANCE
- PLA, PHOSPHOLIPASE A1
- PRI, POSITIVE REGULATOR OF IRON DEFICIENCY RESPONSE
- PSs, phytosiderophores
- PT, peptide transport
- PYE, POPEYE
- RNA sequencing
- SAM, S-adenosyl-L-methionine
- SAMS, S-ADENOSYL-L-METHIONINE SYNTHETASE
- SLC40A1, SOLUTE CARRIER FAMILY 40 MEMBER 1
- SWEET, SUGARS WILL EVENTUALLY BE EXPORTED TRANSPORTERS
- TOM, TRANSPORTER OF MUGINEIC ACID
- Transcriptomic profiles
- VIT, VACUOLAR IRON TRANSPORTER
- Wheat
- YSL, YELLOW STRIPE LIKE
- ZIFL, ZINC INDUCED FACILITATOR-LIKE
- ZIP, ZINC/IRON PERMEASE
- bHLH, BASIC HELIX-LOOP-HELIX
- bZIP, BASIC LEUCINE ZIPPER
- epiHDMA, 3-epihydroxy-2′-deoxymugineic acid
- epiHMA, 3-epihydroxymugineic acid
Collapse
Affiliation(s)
- Meng Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Institute of Molecular Plant Biology, Department of Biology, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| | - Jiazhen Gong
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Navreet K. Bhullar
- Institute of Molecular Plant Biology, Department of Biology, ETH Zurich (Swiss Federal Institute of Technology), Zurich, Switzerland
| |
Collapse
|
20
|
Park KC, Paluncic J, Kovacevic Z, Richardson DR. Pharmacological targeting and the diverse functions of the metastasis suppressor, NDRG1, in cancer. Free Radic Biol Med 2020; 157:154-175. [PMID: 31132412 DOI: 10.1016/j.freeradbiomed.2019.05.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/24/2019] [Accepted: 05/16/2019] [Indexed: 12/18/2022]
Abstract
N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that is regulated by hypoxia, metal ions including iron, the free radical nitric oxide (NO.), and various stress stimuli. This intriguing molecule exhibits diverse functions in cancer, inhibiting epithelial-mesenchymal transition (EMT), cell migration and angiogenesis by modulation of a plethora of oncogenes via cellular signaling. Thus, pharmacological targeting of NDRG1 signaling in cancer is a promising therapeutic strategy. Of note, novel anti-tumor agents of the di-2-pyridylketone thiosemicarbazone series, which exert the "double punch" mechanism by binding metal ions to form redox-active complexes, have been demonstrated to markedly up-regulate NDRG1 expression in cancer cells. This review describes the mechanisms underlying NDRG1 modulation by the thiosemicarbazones and the diverse effects NDRG1 exerts in cancer. As a major induction mechanism, iron depletion appears critical, with NO. also inducing NDRG1 through its ability to bind iron and generate dinitrosyl-dithiol iron complexes, which are then effluxed from cells. Apart from its potent anti-metastatic role, several studies have reported a pro-oncogenic role of NDRG1 in a number of cancer-types. Hence, it has been suggested that NDRG1 plays pleiotropic roles depending on the cancer-type. The molecular mechanism(s) underlying NDRG1 pleiotropy remain elusive, but are linked to differential regulation of WNT signaling and potentially differential interaction with the tumor suppressor, PTEN. This review discusses NDRG1 induction mechanisms by metal ions and NO. and both the anti- and possible pro-oncogenic functions of NDRG1 in multiple cancer-types and compares the opposite effects this protein exerts on cancer progression.
Collapse
Affiliation(s)
- Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Jasmina Paluncic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Zaklina Kovacevic
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Discipline of Pathology and Bosch Institute, Medical Foundation Building (K25), The University of Sydney, Sydney, New South Wales, 2006, Australia.
| |
Collapse
|
21
|
Vanin AF. The Free-Radical Nature of Nitric Oxide Molecules as a Determinant of their Conversion to Nitrosonium Cations in Living Systems. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920030239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
22
|
Zhang X, Wang L, Li H, Zhang L, Zheng X, Cheng W. Crosstalk between noncoding RNAs and ferroptosis: new dawn for overcoming cancer progression. Cell Death Dis 2020; 11:580. [PMID: 32709863 PMCID: PMC7381619 DOI: 10.1038/s41419-020-02772-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
Cancer progression including proliferation, metastasis, and chemoresistance has become a serious hindrance to cancer therapy. This phenomenon mainly derives from the innate insensitive or acquired resistance of cancer cells to apoptosis. Ferroptosis is a newly discovered mechanism of programmed cell death characterized by peroxidation of the lipid membrane induced by reactive oxygen species. Ferroptosis has been confirmed to eliminate cancer cells in an apoptosis-independent manner, however, the specific regulatory mechanism of ferroptosis is still unknown. The use of ferroptosis for overcoming cancer progression is limited. Noncoding RNAs have been found to play an important roles in cancer. They regulate gene expression to affect biological processes of cancer cells such as proliferation, cell cycle, and cell death. Thus far, the functions of ncRNAs in ferroptosis of cancer cells have been examined, and the specific mechanisms by which noncoding RNAs regulate ferroptosis have been partially discovered. However, there is no summary of ferroptosis associated noncoding RNAs and their functions in different cancer types. In this review, we discuss the roles of ferroptosis-associated noncoding RNAs in detail. Moreover, future work regarding the interaction between noncoding RNAs and ferroptosis is proposed, the possible obstacles are predicted and associated solutions are put forward. This review will deepen our understanding of the relationship between noncoding RNAs and ferroptosis, and provide new insights in targeting noncoding RNAs in ferroptosis associated therapeutic strategies.
Collapse
Affiliation(s)
- Xuefei Zhang
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Lingling Wang
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Haixia Li
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China
| | - Lei Zhang
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China.
| | - Xiulan Zheng
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China.
| | - Wen Cheng
- Department of Ultrasonography, Harbin Medical University Cancer Hospital, 150 Haping Road, 150040, Harbin, China.
| |
Collapse
|
23
|
Yanatori I, Richardson DR, Toyokuni S, Kishi F. The new role of poly (rC)-binding proteins as iron transport chaperones: Proteins that could couple with inter-organelle interactions to safely traffic iron. Biochim Biophys Acta Gen Subj 2020; 1864:129685. [PMID: 32679248 DOI: 10.1016/j.bbagen.2020.129685] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intracellular iron transport is mediated by iron chaperone proteins known as the poly(rC)-binding proteins (PCBPs), which were originally identified as RNA/DNA-binding molecules. SCOPE OF REVIEW PCBPs assume a role as not only as cytosolic iron carriers, but also as regulators of iron transport and recycling. PCBP1 is involved in the iron storage pathway that involves ferritin, while PCBP2 is involved in processes that include: iron transfer from the iron importer, divalent metal ion transporter 1; iron export mediated by ferroportin-1; and heme degradation via heme oxygenase 1. MAJOR CONCLUSIONS Both PCBP1 and PCBP2 possess iron-binding activity and form hetero/homo dimer complexes. These iron chaperones have a subset of non-redundant functions and regulate iron metabolism independently. GENERAL SIGNIFICANCE This intracellular iron chaperone system mediated by PCBPs provide a transport "gateway" of ferrous iron that may potentially link with dynamic, inter-organelle interactions to safely traffic intracellular iron.
Collapse
Affiliation(s)
- Izumi Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Des R Richardson
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Centre for Cancer Cell Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Queensland, Australia
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | |
Collapse
|
24
|
El-Garawani IM, El-Nabi SH, El-Shafey S, Elfiky M, Nafie E. Coffea arabica Bean Extracts and Vitamin C: A Novel Combination Unleashes MCF-7 Cell Death. Curr Pharm Biotechnol 2020; 21:23-36. [PMID: 31438827 DOI: 10.2174/1389201020666190822161337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/13/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Vitamin C (VC) is believed to enhance immunity and is regularly integrated as a supplementary agent during several treatments. OBJECTIVE The green (GC) and roasted (RC) coffee (Coffea arabica) aqueous extracts (0, 125, 250 and 500 μg/ml) combined with VC (50 μg/ml) were examined on the cancerous MCF-7 cell line and normal human lymphocytes. METHODS Neutral red uptake assay, comet assay, immunocytochemical reactivity for protein expression and mRNA expression of apoptosis-related genes were performed. RESULTS A significant (P< 0.05) concentration-dependent increase of apoptotic features, such as morphological changes, and abundant nuclear condensation, altered the expression of p53 and caspase-3 mRNA, down-regulation of Bcl-2 protein as well as the acidic autophagosomal vacuolization in treated cells. The oxidative stress and DNA single-strand breaks were noticed too. CONCLUSION These results suggest that coffee in combination with VC undergoes apoptotic anticancer pathway. This supports the integration of coffee and VC as a valuable candidate for anticancer research and treatments.
Collapse
Affiliation(s)
- Islam M El-Garawani
- Zoology Department, Faculty of Science, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Sobhy H El-Nabi
- Zoology Department, Faculty of Science, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Samraa El-Shafey
- Zoology Department, Faculty of Science, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mohamed Elfiky
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Ebtesam Nafie
- Department of Zoology, Faculty of Science, Benha University, Benha, Egypt
| |
Collapse
|
25
|
Winn NC, Volk KM, Hasty AH. Regulation of tissue iron homeostasis: the macrophage "ferrostat". JCI Insight 2020; 5:132964. [PMID: 31996481 DOI: 10.1172/jci.insight.132964] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Iron is an essential element for multiple fundamental biological processes required for life; yet iron overload can be cytotoxic. Consequently, iron concentrations at the cellular and tissue level must be exquisitely governed by mechanisms that complement and fine-tune systemic control. It is well appreciated that macrophages are vital for systemic iron homeostasis, supplying or sequestering iron as needed for erythropoiesis or bacteriostasis, respectively. Indeed, recycling of iron through erythrophagocytosis by splenic macrophages is a major contributor to systemic iron homeostasis. However, accumulating evidence suggests that tissue-resident macrophages regulate local iron availability and modulate the tissue microenvironment, contributing to cellular and tissue function. Here, we summarize the significance of tissue-specific regulation of iron availability and highlight how resident macrophages are critical for this process. This tissue-dependent regulation has broad implications for understanding both resident macrophage function and tissue iron homeostasis in health and disease.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Katrina M Volk
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
26
|
Cho SL, Liao CJ, Lu TT. Synthetic methodology for preparation of dinitrosyl iron complexes. J Biol Inorg Chem 2019; 24:495-515. [DOI: 10.1007/s00775-019-01668-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/15/2019] [Indexed: 12/29/2022]
|
27
|
Wiese M, Stefan SM. The A‐B‐C of small‐molecule ABC transport protein modulators: From inhibition to activation—a case study of multidrug resistance‐associated protein 1 (ABCC1). Med Res Rev 2019; 39:2031-2081. [DOI: 10.1002/med.21573] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Michael Wiese
- Pharmaceutical Institute, Rheinische Friedrich‐Wilhelms‐University of Bonn Bonn Germany
| | - Sven Marcel Stefan
- Pharmaceutical Institute, Rheinische Friedrich‐Wilhelms‐University of Bonn Bonn Germany
| |
Collapse
|
28
|
Skaf M, Hany S, Aouad S, Labaki M, Abi-Aad E, Aboukaïs A. Adsorption of probe molecules to investigate by EPR the redox properties of silver loaded on ceria. Chem Phys 2019. [DOI: 10.1016/j.chemphys.2018.10.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
29
|
Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol Res 2019; 139:298-313. [DOI: 10.1016/j.phrs.2018.11.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
30
|
Hsiao HY, Chung CW, Santos JH, Villaflores OB, Lu TT. Fe in biosynthesis, translocation, and signal transduction of NO: toward bioinorganic engineering of dinitrosyl iron complexes into NO-delivery scaffolds for tissue engineering. Dalton Trans 2019; 48:9431-9453. [DOI: 10.1039/c9dt00777f] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ubiquitous physiology of nitric oxide enables the bioinorganic engineering of [Fe(NO)2]-containing and NO-delivery scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Center for Tissue Engineering
- Chang Gung Memorial Hospital
- Taoyuan
- Taiwan
| | - Chieh-Wei Chung
- Institute of Biomedical Engineering
- National Tsing Hua University
- Hsinchu
- Taiwan
| | | | - Oliver B. Villaflores
- Department of Biochemistry
- Faculty of Pharmacy
- University of Santo Tomas
- Manila
- Philippines
| | - Tsai-Te Lu
- Institute of Biomedical Engineering
- National Tsing Hua University
- Hsinchu
- Taiwan
| |
Collapse
|
31
|
Lu TT, Wang YM, Hung CH, Chiou SJ, Liaw WF. Bioinorganic Chemistry of the Natural [Fe(NO)2] Motif: Evolution of a Functional Model for NO-Related Biomedical Application and Revolutionary Development of a Translational Model. Inorg Chem 2018; 57:12425-12443. [DOI: 10.1021/acs.inorgchem.8b01818] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Yun-Ming Wang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30013, Taiwan
| | | | - Show-Jen Chiou
- Department of Applied Chemistry, National Chiayi University, Chiayi 60004, Taiwan
| | | |
Collapse
|
32
|
Mukosera GT, Liu T, Ishtiaq Ahmed AS, Li Q, Sheng MHC, Tipple TE, Baylink DJ, Power GG, Blood AB. Detection of dinitrosyl iron complexes by ozone-based chemiluminescence. Nitric Oxide 2018; 79:57-67. [PMID: 30059767 PMCID: PMC6277231 DOI: 10.1016/j.niox.2018.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Dinitrosyl iron complexes (DNICs) are important intermediates in the metabolism of nitric oxide (NO). They have been considered to be NO storage adducts able to release NO, scavengers of excess NO during inflammatory hypotensive shock, and mediators of apoptosis in cancer cells, among many other functions. Currently, all studies of DNICs in biological matrices use electron paramagnetic resonance (EPR) for both detection and quantification. EPR is limited, however, by its ability to detect only paramagnetic mononuclear DNICs even though EPR-silent binuclear are likely to be prevalent. Furthermore, physiological concentrations of mononuclear DNICs are usually lower than the EPR detection limit (1 μM). We have thus developed a chemiluminescence-based method for the selective detection of both DNIC forms at physiological, pathophysiological, and pharmacologic conditions. We have also demonstrated the use of the new method in detecting DNIC formation in the presence of nitrite and nitrosothiols within biological fluids and tissue. This new method, which can be used alone or in tandem with EPR, has the potential to offer insight about the involvement of DNICs in many NO-dependent pathways.
Collapse
Affiliation(s)
- George T Mukosera
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Taiming Liu
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Abu Shufian Ishtiaq Ahmed
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Center for Dental Research, Loma Linda University School of Dentistry, Loma Linda, CA, 92350, USA
| | - Qian Li
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Matilda H-C Sheng
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David J Baylink
- Regenerative Medicine Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Gordon G Power
- Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Arlin B Blood
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA; Lawrence D. Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
33
|
Lewandowska H, Stępkowski TM, Męczyńska-Wielgosz S, Sikorska K, Sadło J, Dudek J, Kruszewski M. LDL dinitrosyl iron complex acts as an iron donor in mouse macrophages. J Inorg Biochem 2018; 188:29-37. [PMID: 30119015 DOI: 10.1016/j.jinorgbio.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/22/2018] [Accepted: 08/02/2018] [Indexed: 12/27/2022]
Abstract
[Fe(NO)2] - modified nanoparticles of low-density protein (DNICLDL) can serve as conveyors of iron in the form of stable complexes with ApoB100 protein. As reported recently, in human hepatoma cells DNICLDL significantly increased the total iron content, while showing low toxicity. In the present work, we focused on the effects of internalization of DNIC-modified lipoproteins in macrophages, with special regards to cytotoxicity. DNICLDL was administered to a model macrophage cell line, RAW 264.7. Administration of DNICLDL considerably increased total iron content. High increase of iron was accompanied by moderate toxicity. As shown by in vitro plasmid nicking assay, chelation of iron in the form of DNIC strongly reduced the iron-related reactive oxygen species (ROS) -induced DNA damage. In addition, DNICLDL, plausibly due to its NO-donating activity, did not induce inducible nitric oxide synthase (iNOS) expression, as opposed to other forms of low-density protein (LDL).
Collapse
Affiliation(s)
- Hanna Lewandowska
- Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland.
| | - Tomasz M Stępkowski
- Laboratory of Mitochondrial Biogenesis, Centre of New Technologies UW, Banacha 2c, 02-097 Warsaw, Poland
| | | | - Katarzyna Sikorska
- Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland
| | - Jarosław Sadło
- Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland
| | - Jakub Dudek
- Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, 16 Dorodna Str., 03-195 Warsaw, Poland; Faculty of Medicine, University of Information Technology and Management in Rzeszów, ul. Sucharskiego 2, 35-225 Rzeszów, Poland; Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| |
Collapse
|
34
|
Stefan SM, Wiese M. Small-molecule inhibitors of multidrug resistance-associated protein 1 and related processes: A historic approach and recent advances. Med Res Rev 2018; 39:176-264. [DOI: 10.1002/med.21510] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sven Marcel Stefan
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| | - Michael Wiese
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| |
Collapse
|
35
|
Sahni S, Hickok JR, Thomas DD. Nitric oxide reduces oxidative stress in cancer cells by forming dinitrosyliron complexes. Nitric Oxide 2018. [PMID: 29522907 DOI: 10.1016/j.niox.2018.03.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The chelatable iron pool (CIP) is a small but chemically significant fraction of total cellular iron. While this dynamic population of iron is limited, it is redox active and capable of generating reactive oxygen species (ROS) that can lead to oxidative stress which is associated with various pathologies. Nitric oxide (•NO), is a free radical signalling molecule that regulates numerous physiological and pathological conditions. We have previously shown that macrophages exposed to endogenously generated or exogenously administered nitric oxide (•NO) results in its interaction with CIP to form dinitrosyliron complexes with thiol containing ligands (DNICs). In this study we assessed the consequences of DNIC formation in cancer cells as •NO is known to be associated with numerous malignancies. Incubation of cancer cells with •NO led to a time and dose dependent increase in formation of DNICs. The formation of DNICs results in the sequestration of the CIP which is a major source of iron for redox reactions and reactive oxygen species (ROS) generation. Therefore, we set out to test the antioxidant effect of •NO by measuring the ability of DNICs to protect cells against oxidative stress. We observed that cancer cells treated with •NO were partially protected against H2O2 mediated cytotoxicity. This correlated to a concomitant decrease in the formation of oxidants when •NO was present during H2O2 treatment. Similar protective effects were achieved by treating cells with iron chelators in the presence of H2O2. Interestingly, •NO decreased the rate of cellular metabolism of H2O2 suggesting that a proportion of H2O2 is consumed via reactions with cellular iron. When the CIP was artificially increased by supplementation of cells with iron, a significant decrease in the cytoprotective effect of •NO was observed. Notably, •NO concentrations, at which cytoprotective and antioxidant effects were observed, correlated with concentration-dependent increases in DNIC formation. Collectively, these results demonstrate that •NO has antioxidant properties by its ability to sequester cellular iron. This could play a significant role in variety of diseases involving ROS mediated toxicity like cancer and neurodegenerative disorders where •NO has been shown to be an important etiologic factor.
Collapse
Affiliation(s)
- Sumit Sahni
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Australia
| | - Jason R Hickok
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Douglas D Thomas
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
36
|
Abstract
SIGNIFICANCE Glutathione (GSH) is the most abundant cellular low-molecular-weight thiol in the majority of organisms in all kingdoms of life. Therefore, functions of GSH and disturbed regulation of its concentration are associated with numerous physiological and pathological situations. Recent Advances: The function of GSH as redox buffer or antioxidant is increasingly being questioned. New functions, especially functions connected to the cellular iron homeostasis, were elucidated. Via the formation of iron complexes, GSH is an important player in all aspects of iron metabolism: sensing and regulation of iron levels, iron trafficking, and biosynthesis of iron cofactors. The variety of GSH coordinated iron complexes and their functions with a special focus on FeS-glutaredoxins are summarized in this review. Interestingly, GSH analogues that function as major low-molecular-weight thiols in organisms lacking GSH resemble the functions in iron homeostasis. CRITICAL ISSUES Since these iron-related functions are most likely also connected to thiol redox chemistry, it is difficult to distinguish between mechanisms related to either redox or iron metabolisms. FUTURE DIRECTIONS The ability of GSH to coordinate iron in different complexes with or without proteins needs further investigation. The discovery of new Fe-GSH complexes and their physiological functions will significantly advance our understanding of cellular iron homeostasis. Antioxid. Redox Signal. 27, 1235-1251.
Collapse
Affiliation(s)
- Carsten Berndt
- 1 Department of Neurology, Medical Faculty, Life Science Center , Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Christopher Horst Lillig
- 2 Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald , Greifswald, Germany
| |
Collapse
|
37
|
Vanin AF. Dinitrosyl iron complexes with thiol-containing ligands as a base for developing drugs with diverse therapeutic activities: Physicochemical and biological substantiation. Biophysics (Nagoya-shi) 2017. [DOI: 10.1134/s0006350917040224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
38
|
Kazan HH, Urfali-Mamatoglu C, Gunduz U. Iron metabolism and drug resistance in cancer. Biometals 2017; 30:629-641. [DOI: 10.1007/s10534-017-0037-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/26/2017] [Indexed: 01/17/2023]
|
39
|
Toyokuni S, Ito F, Yamashita K, Okazaki Y, Akatsuka S. Iron and thiol redox signaling in cancer: An exquisite balance to escape ferroptosis. Free Radic Biol Med 2017; 108:610-626. [PMID: 28433662 DOI: 10.1016/j.freeradbiomed.2017.04.024] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Epidemiological data indicate a constant worldwide increase in cancer mortality, although the age of onset is increasing. Recent accumulation of genomic data on human cancer via next-generation sequencing confirmed that cancer is a disease of genome alteration. In many cancers, the Nrf2 transcription system is activated via mutations either in Nrf2 or Keap1 ubiquitin ligase, leading to persistent activation of the genes with antioxidative functions. Furthermore, deep sequencing of passenger mutations is clarifying responsible cancer causative agent(s) in each case, including aging, APOBEC activation, smoking and UV. Therefore, it is most likely that oxidative stress is the principal initiating factor in carcinogenesis, with the involvement of two essential molecules for life, iron and oxygen. There is evidence based on epidemiological and animal studies that excess iron is a major risk for carcinogenesis, suggesting the importance of ferroptosis-resistance. Microscopic visualization of catalytic Fe(II) has recently become available. Although catalytic Fe(II) is largely present in lysosomes, proliferating cells harbor catalytic Fe(II) also in the cytosol and mitochondria. Oxidative stress catalyzed by Fe(II) is counteracted by thiol systems at different functional levels. Nitric oxide, carbon monoxide and hydrogen (per)sulfide modulate these reactions. Mitochondria generate not only energy but also heme/iron sulfur cluster cofactors and remain mostly dysfunctional in cancer cells, leading to Warburg effects. Cancer cells are under persistent oxidative stress with a delicate balance between catalytic iron and thiols, thereby escaping ferroptosis. Thus, high-dose L-ascorbate and non-thermal plasma as well as glucose/glutamine deprivation may provide additional benefits as cancer therapies over preexisting therapeutics.
Collapse
Affiliation(s)
- Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Sydney Medical School, The University of Sydney, NSW 2006, Australia.
| | - Fumiya Ito
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kyoko Yamashita
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yasumasa Okazaki
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinya Akatsuka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
40
|
Lepka K, Volbracht K, Bill E, Schneider R, Rios N, Hildebrandt T, Ingwersen J, Prozorovski T, Lillig CH, van Horssen J, Steinman L, Hartung HP, Radi R, Holmgren A, Aktas O, Berndt C. Iron-sulfur glutaredoxin 2 protects oligodendrocytes against damage induced by nitric oxide release from activated microglia. Glia 2017; 65:1521-1534. [PMID: 28618115 DOI: 10.1002/glia.23178] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
Demyelinated brain lesions, a hallmark of autoimmune neuroinflammatory diseases like multiple sclerosis, result from oligodendroglial cell damage. Activated microglia are considered a major source of nitric oxide and subsequent peroxynitrite-mediated damage of myelin. Here, we provide biochemical and biophysical evidence that the oxidoreductase glutaredoxin 2 inhibits peroxynitrite formation by transforming nitric oxide into dinitrosyl-diglutathionyl-iron-complexes. Glutaredoxin 2 levels influence both survival rates of primary oligodendrocyte progenitor cells and preservation of myelin structure in cerebellar organotypic slice cultures challenged with activated microglia or nitric oxide donors. Of note, glutaredoxin 2-mediated protection is not linked to its enzymatic activity as oxidoreductase, but to the disassembly of its uniquely coordinated iron-sulfur cluster using glutathione as non-protein ligand. The protective effect of glutaredoxin 2 is connected to decreased protein carbonylation and nitration. In line, brain lesions of mice suffering from experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, show decreased glutaredoxin 2 expression and increased nitrotyrosine formation indicating that this type of protection is missing in the inflamed central nervous system. Our findings link inorganic biochemistry to neuroinflammation and identify glutaredoxin 2 as a protective factor against neuroinflammation-mediated myelin damage. Thus, improved availability of glutathione-coordinated iron-sulfur clusters emerges as a potential therapeutic approach in inflammatory demyelination.
Collapse
Affiliation(s)
- Klaudia Lepka
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Katrin Volbracht
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Eckhard Bill
- Max-Planck-Institut für Chemische Energiekonversion, Mülheim/Ruhr, 45470, Germany
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Natalia Rios
- Departmento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, CP 11800, Uruguay
| | - Thomas Hildebrandt
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Timur Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Christopher Horst Lillig
- Universitätsmedizin Greifswald, Institute for Medical Biochemistry and Molecular Biology, Greifswald, 17475, Germany
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, MB, 1007, The Netherlands
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, 94305-5316, USA
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Rafael Radi
- Departmento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, CP 11800, Uruguay
| | - Arne Holmgren
- Department for Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine Universität, Düsseldorf, 40225, Germany
| |
Collapse
|
41
|
Keszler A, Diers AR, Ding Z, Hogg N. Thiolate-based dinitrosyl iron complexes: Decomposition and detection and differentiation from S-nitrosothiols. Nitric Oxide 2017; 65:1-9. [PMID: 28111306 PMCID: PMC5663227 DOI: 10.1016/j.niox.2017.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/22/2016] [Accepted: 01/18/2017] [Indexed: 11/26/2022]
Abstract
Dinitrosyl iron complexes (DNIC) spontaneously form in aqueous solutions of Fe(II), nitric oxide (NO), and various anions. They exist as an equilibrium between diamagnetic, dimeric (bi-DNIC) and paramagnetic, monomeric (mono-DNIC) forms. Thiolate groups (e.g., on glutathione or protein cysteine residues) are the most biologically relevant anions to coordinate to Fe(II). Low molecular weight DNIC have previously been suggested to be important mediators of NO biology in cells, and emerging literature supports their role in the control of iron-dependent cellular processes. Recently, it was shown that DNIC may be one of the most abundant NO-derived products in cells and may serve as intermediates in the cellular formation of S-nitrosothiols. In this work, we examined the stability of low molecular weight DNIC and investigated issues with their detection in the presence of other NO-dependent metabolites such as S-nitrosothiols. By using spectrophotometric, Electron Paramagnetic Resonance, ozone-based chemiluminesence, and HPLC techniques we established that at neutral pH, bi-DNIC remain stable for hours, whereas excess thiol results in decomposition to form nitrite. NO was also detected during the decomposition, but no S-nitrosothiol formation was observed. Importantly, mercury chloride accelerated the degradation of DNIC; thus, the implications of this finding for the diagnostic use of mercury chloride in the detection of S-nitrosothiols were determined in simple and complex biological systems. We conclude S-nitrosothiol levels may have been substantially overestimated in all methods where mercury chloride has been used.
Collapse
Affiliation(s)
- Agnes Keszler
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Anne R Diers
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Zhen Ding
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Neil Hogg
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, Milwaukee, WI 53226, United States.
| |
Collapse
|
42
|
Regulation and control of nitric oxide (NO) in macrophages: Protecting the “professional killer cell” from its own cytotoxic arsenal via MRP1 and GSTP1. Biochim Biophys Acta Gen Subj 2017; 1861:995-999. [DOI: 10.1016/j.bbagen.2017.02.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/14/2017] [Indexed: 11/22/2022]
|
43
|
Vanin AF, Borodulin RR, Mikoyan VD. Dinitrosyl iron complexes with natural thiol-containing ligands in aqueous solutions: Synthesis and some physico-chemical characteristics (A methodological review). Nitric Oxide 2017; 66:1-9. [PMID: 28216238 DOI: 10.1016/j.niox.2017.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 01/16/2023]
Abstract
Two approaches to the synthesis of dinitrosyl iron complexes (DNIC) with glutathione and l-cysteine in aqueous solutions based on the use of gaseous NO and appropriate S-nitrosothiols, viz., S-nitrosoglutathione (GS-NO) or S-nitrosocysteine (Cys-NO), respectively, are considered. A schematic representation of a vacuum unit for generation and accumulation of gaseous NO purified from the NO2 admixture and its application for obtaining aqueous solutions of DNIC in a Thunberg apparatus is given. To achieve this, a solution of bivalent iron in distilled water is loaded into the upper chamber of the Thunberg apparatus, while the thiol solution in an appropriate buffer (рН 7.4) is loaded into its lower chamber. Further steps, which include degassing, addition of gaseous NO, shaking of both solutions and formation of the Fe2+-thiol mixture, culminate in the synthesis of DNIC. The second approach consists in a stepwise addition of Fe2+ salts and nitrite to aqueous solutions of glutathione or cysteine. In the presence of Fe2+ and after the increase in рН to the physiological level, GS-NO or Cys-NO generated at acid media (pH < 4) are converted into DNIC with glutathione or cysteine. Noteworthy, irrespective of the procedure used for their synthesis DNIC with glutathione manifest much higher stability than DNIC with cysteine. The pattern of spin density distribution in iron-dinitrosyl fragments of DNIC characterized by the d7 electronic configuration of the iron atom and described by the formula Fe+(NO+)2 is unique in that it provides a plausible explanation for the ability of DNIC to generate NO and nitrosonium ions (NO+) and the peculiar characteristics of the EPR signal of their mononuclear form (M-DNIC).
Collapse
Affiliation(s)
- Anatoly F Vanin
- N.N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Institute of Regenerative Medicine, I.M. Sechenov First Moscow Medical University, Moscow, Russia.
| | - Rostislav R Borodulin
- N.N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Vasak D Mikoyan
- N.N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
44
|
Timoshin AA, Lakomkin VL, Abramov AA, Ruuge EK, Vanin AF. Hypotensive Effect and Accumulation of Dinitrosyl Iron Complexes in Blood and Tissues after Intravenous and Subcutaneous Injection. Bull Exp Biol Med 2016; 162:207-210. [PMID: 27913937 DOI: 10.1007/s10517-016-3577-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Indexed: 10/20/2022]
Abstract
Subcutaneous injection of Oxacom with glutathione-bound dinitrosyl iron complex as the active principle produced a slower drop of mean BP and longer accumulation of protein-bound dinitrosyl iron complexes in whole blood and tissues than intravenous injection of this drug, while durations of hypotensive effect in both cases were practically identical. In contrast to intravenous injection of the drug, its subcutaneous administration was not characterized by a high concentration of protein-bound dinitrosyl iron complexes in the blood at the onset of experiment; in addition, accumulation of these NO forms in the lungs was more pronounced after subcutaneous injection than after intravenous one.
Collapse
Affiliation(s)
- A A Timoshin
- Research Institute of Experimental Cardiology of Russian Cardiology Research and Production Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V L Lakomkin
- Research Institute of Experimental Cardiology of Russian Cardiology Research and Production Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A A Abramov
- Research Institute of Experimental Cardiology of Russian Cardiology Research and Production Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E K Ruuge
- Research Institute of Experimental Cardiology of Russian Cardiology Research and Production Complex, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A F Vanin
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
45
|
Lok HC, Sahni S, Jansson PJ, Kovacevic Z, Hawkins CL, Richardson DR. A Nitric Oxide Storage and Transport System That Protects Activated Macrophages from Endogenous Nitric Oxide Cytotoxicity. J Biol Chem 2016; 291:27042-27061. [PMID: 27866158 DOI: 10.1074/jbc.m116.763714] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/16/2016] [Indexed: 12/20/2022] Open
Abstract
Nitric oxide (NO) is integral to macrophage cytotoxicity against tumors due to its ability to induce iron release from cancer cells. However, the mechanism for how activated macrophages protect themselves from endogenous NO remains unknown. We previously demonstrated by using tumor cells that glutathione S-transferase P1 (GSTP1) sequesters NO as dinitrosyl-dithiol iron complexes (DNICs) and inhibits NO-mediated iron release from cells via the transporter multidrug resistance protein 1 (MRP1/ABCC1). These prior studies also showed that MRP1 and GSTP1 protect tumor cells against NO cytotoxicity, which parallels their roles in defending cancer cells from cytotoxic drugs. Considering this, and because GSTP1 and MRP1 are up-regulated during macrophage activation, this investigation examined whether this NO storage/transport system protects macrophages against endogenous NO cytotoxicity in two well characterized macrophage cell types (J774 and RAW 264.7). MRP1 expression markedly increased upon macrophage activation, and the role of MRP1 in NO-induced 59Fe release was demonstrated by Mrp1 siRNA and the MRP1 inhibitor, MK571, which inhibited NO-mediated iron efflux. Furthermore, Mrp1 silencing increased DNIC accumulation in macrophages, indicating a role for MRP1 in transporting DNICs out of cells. In addition, macrophage 59Fe release was enhanced by silencing Gstp1, suggesting GSTP1 was responsible for DNIC binding/storage. Viability studies demonstrated that GSTP1 and MRP1 protect activated macrophages from NO cytotoxicity. This was confirmed by silencing nuclear factor-erythroid 2-related factor 2 (Nrf2), which decreased MRP1 and GSTP1 expression, concomitant with reduced 59Fe release and macrophage survival. Together, these results demonstrate a mechanism by which macrophages protect themselves against NO cytotoxicity.
Collapse
Affiliation(s)
- Hiu Chuen Lok
- From the Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Sumit Sahni
- From the Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Patric J Jansson
- From the Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Zaklina Kovacevic
- From the Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Clare L Hawkins
- the Heart Research Institute, Sydney, New South Wales 2042, Australia
| | - Des R Richardson
- From the Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| |
Collapse
|
46
|
Mikoyan VD, Burgova EN, Borodulin RR, Vanin AF. The binuclear form of dinitrosyl iron complexes with thiol-containing ligands in animal tissues. Nitric Oxide 2016; 62:1-10. [PMID: 27989818 DOI: 10.1016/j.niox.2016.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/19/2016] [Accepted: 10/27/2016] [Indexed: 11/18/2022]
Abstract
It has been established that treatment of mice with sodium nitrite, S-nitrosoglutathione and the water-soluble nitroglycerine derivative isosorbide dinitrate (ISDN) as NO donors initiates in vivo synthesis of significant amounts of EPR-silent binuclear dinitrosyl iron complexes (B-DNIC) with thiol-containing ligands in the liver and other tissues of experimental mice. This effect is especially apparent if NO donors are administered to mice simultaneously with the Fe2+-citrate complex. Similar results were obtained in experiments on isolated liver and other mouse tissues treated with gaseous NО in vitro and during stimulation of endogenous NO synthesis in the presence of inducible NO synthase. B-DNIC appeared in mouse tissues after in vitro treatment of tissue samples with an aqueous solution of diethyldithiocarbamate (DETC), which resulted in the transfer of iron-mononitrosyl fragments from B-DNIC to the thiocarbonyl group of DETC and the formation of EPR-detectable mononitrosyl iron complexes (MNIC) with DETC. EPR-Active MNIC with N-methyl-d-glucamine dithiocarbamate (MGD) were synthesized in a similar way. MNIC-MGD were also formed in the reaction of water-soluble MGD-Fe2+ complexes with sodium nitrite, S-nitrosoglutathione and ISDN.
Collapse
Affiliation(s)
- Vasak D Mikoyan
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Evgeniya N Burgova
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Rostislav R Borodulin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly F Vanin
- N.N.Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russia; Institute for Regenerative Medicine, I.M.Sechenov Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
47
|
Lancaster JR. How are nitrosothiols formed de novo in vivo? Arch Biochem Biophys 2016; 617:137-144. [PMID: 27794428 DOI: 10.1016/j.abb.2016.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 10/23/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
The biological mechanisms of de novo formation of cellular nitrosothiols (as opposed to transnitrosation) are reviewed. The approach is to introduce chemical foundations for each mechanism, followed by evidence in biological systems. The general categories include mechanisms involving nitrous acid, NO autoxidation and oxidant stress, redox active and inactive metal ions, and sulfide/persulfide. Important conclusions/speculations are that de novo cellular thiol nitrosation (1) is an oxidative process, and so should be considered within the family of other thiol oxidative modifications, (2) may not involve a single dominant process but depends on the specific conditions, (3) does not involve O2 under at least some conditions, and (4) may serve to provide a "substrate pool" of protein cysteine nitrosothiol which could, through subsequent enzymatic transnitrosation/denitrosation, be "rearranged" to accomplish the specificity and regulatory control required for effective post-translational signaling.
Collapse
Affiliation(s)
- Jack R Lancaster
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, United States; Department of Medicine, University of Pittsburgh School of Medicine, United States; Department of Surgery, University of Pittsburgh School of Medicine, United States
| |
Collapse
|
48
|
Lui GYL, Kovacevic Z, Richardson V, Merlot AM, Kalinowski DS, Richardson DR. Targeting cancer by binding iron: Dissecting cellular signaling pathways. Oncotarget 2016; 6:18748-79. [PMID: 26125440 PMCID: PMC4662454 DOI: 10.18632/oncotarget.4349] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/12/2015] [Indexed: 12/30/2022] Open
Abstract
Newer and more potent therapies are urgently needed to effectively treat advanced cancers that have developed resistance and metastasized. One such strategy is to target cancer cell iron metabolism, which is altered compared to normal cells and may facilitate their rapid proliferation. This is supported by studies reporting the anti-neoplastic activities of the clinically available iron chelators, desferrioxamine and deferasirox. More recently, ligands of the di-2-pyridylketone thiosemicarbazone (DpT) class have demonstrated potent and selective anti-proliferative activity across multiple cancer-types in vivo, fueling studies aimed at dissecting their molecular mechanisms of action. In the past five years alone, significant advances have been made in understanding how chelators not only modulate cellular iron metabolism, but also multiple signaling pathways implicated in tumor progression and metastasis. Herein, we discuss recent research on the targeting of iron in cancer cells, with a focus on the novel and potent DpT ligands. Several key studies have revealed that iron chelation can target the AKT, ERK, JNK, p38, STAT3, TGF-β, Wnt and autophagic pathways to subsequently inhibit cellular proliferation, the epithelial-mesenchymal transition (EMT) and metastasis. These developments emphasize that these novel therapies could be utilized clinically to effectively target cancer.
Collapse
Affiliation(s)
- Goldie Y L Lui
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Zaklina Kovacevic
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Vera Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Angelica M Merlot
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Danuta S Kalinowski
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Des R Richardson
- Department of Pathology and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
49
|
Modulation of Tamoxifen Cytotoxicity by Caffeic Acid Phenethyl Ester in MCF-7 Breast Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3017108. [PMID: 26697130 PMCID: PMC4677239 DOI: 10.1155/2016/3017108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/27/2015] [Indexed: 12/14/2022]
Abstract
Although Tamoxifen (TAM) is one of the most widely used drugs in managing breast cancer, many women still relapse after long-term therapy. Caffeic acid phenethyl ester (CAPE) is a polyphenolic compound present in many medicinal plants and in propolis. The present study examined the effect of CAPE on TAM cytotoxicity in MCF-7 cells. MCF-7 cells were treated with different concentrations of TAM and/or CAPE for 48 h. This novel combination exerted synergistic cytotoxic effects against MCF-7 cells via induction of apoptotic machinery with activation of caspases and DNA fragmentation, along with downregulation of Bcl-2 and Beclin 1 expression levels. However, the mammalian microtubule-associated protein light chain LC 3-II level was unchanged. Vascular endothelial growth factor level was also decreased, whereas levels of glutathione and nitric oxide were increased. In conclusion, CAPE augmented TAM cytotoxicity via multiple mechanisms, providing a novel therapeutic approach for breast cancer treatment that can overcome resistance and lower toxicity. This effect provides a rationale for further investigation of this combination.
Collapse
|
50
|
Vanin AF, Mikoyan VD, Kubrina LN, Borodulin RR, Burgova EN. Mono- and binuclear dinitrosyl iron complexes with thiol-containing ligands in various biosystems. Biophysics (Nagoya-shi) 2015. [DOI: 10.1134/s0006350915040247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|