1
|
Quadros Barsé L, Ulfig A, Varatnitskaya M, Vázquez-Hernández M, Yoo J, Imann AM, Lupilov N, Fischer M, Becker K, Bandow JE, Leichert LI. Comparison of the mechanism of antimicrobial action of the gold(I) compound auranofin in Gram-positive and Gram-negative bacteria. Microbiol Spectr 2024; 12:e0013824. [PMID: 39377597 PMCID: PMC11537011 DOI: 10.1128/spectrum.00138-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 10/09/2024] Open
Abstract
While highly effective at killing Gram-positive bacteria, auranofin lacks significant activity against Gram-negative species for reasons that largely remain unclear. Here, we aimed to elucidate the molecular mechanisms underlying the low susceptibility of the Gram-negative model organism Escherichia coli to auranofin when compared to the Gram-positive model organism Bacillus subtilis. The proteome response of E. coli exposed to auranofin suggests a combination of inactivation of thiol-containing enzymes and the induction of systemic oxidative stress. Susceptibility tests in E. coli mutants lacking proteins upregulated upon auranofin treatment suggested that none of them are directly involved in E. coli's high tolerance to auranofin. E. coli cells lacking the efflux pump component TolC were more sensitive to auranofin treatment, but not to an extent that would fully explain the observed difference in susceptibility of Gram-positive and Gram-negative organisms. We thus tested whether E. coli's thioredoxin reductase (TrxB) is inherently less sensitive to auranofin than TrxB from B. subtilis, which was not the case. However, E. coli strains lacking the low-molecular-weight thiol glutathione, but not glutathione reductase, showed a high susceptibility to auranofin. Bacterial cells expressing the genetically encoded redox probe roGFP2 allowed us to observe the oxidation of cellular protein thiols in situ. Based on our findings, we hypothesize that auranofin leads to a global disturbance in the cellular thiol redox homeostasis in bacteria, but Gram-negative bacteria are inherently more resistant due to the presence of drug export systems and high cellular concentrations of glutathione.IMPORTANCEAuranofin is an FDA-approved drug for the treatment of rheumatoid arthritis. However, it has also high antibacterial activity, in particular against Gram-positive organisms. In the current antibiotics crisis, this would make it an ideal candidate for drug repurposing. However, its much lower activity against Gram-negative organisms prevents its broad-spectrum application. Here we show that, on the level of the presumed target, there is no difference in susceptibility between Gram-negative and Gram-positive species: thioredoxin reductases from both Escherichia coli and Bacillus subtilis are equally inhibited by auranofin. In both species, auranofin treatment leads to oxidative protein modification on a systemic level, as monitored by proteomics and the genetically encoded redox probe roGFP2. The single largest contributor to E. coli's relative resistance to auranofin seems to be the low-molecular-weight thiol glutathione, which is absent in B. subtilis and other Gram-positive species.
Collapse
Affiliation(s)
- Laísa Quadros Barsé
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Agnes Ulfig
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Marharyta Varatnitskaya
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | | | - Jihyun Yoo
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Astrid M. Imann
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
- Institute of Electrical Engineering and Applied Sciences–Molecular Biology, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - Natalie Lupilov
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Marina Fischer
- Interdisciplinary Research Center, Justus Liebig University Giessen, Giessen, Germany
| | - Katja Becker
- Interdisciplinary Research Center, Justus Liebig University Giessen, Giessen, Germany
| | - Julia E. Bandow
- Faculty of Biology and Biotechnology, Applied Microbiology, Ruhr University Bochum, Bochum, Germany
| | - Lars I. Leichert
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
2
|
El Hanafi K, Fernández-Bautista T, Ouerdane L, Corns WT, Bueno M, Fontagné-Dicharry S, Amouroux D, Pedrero Z. Exploring mercury detoxification in fish: The role of selenium from tuna byproduct diets for sustainable aquaculture. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135779. [PMID: 39298964 DOI: 10.1016/j.jhazmat.2024.135779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
Exposure to mercury (Hg) through fish consumption poses significant environmental and public health risks, given its status as one of the top ten hazardous chemicals. Aquaculture is expanding, driving a surge in demand for sustainable aquafeeds. Tuna byproducts, which are rich in protein, offer potential for aquafeed production, yet their use is challenged by the high content of heavy metals, particularly Hg. However, these byproducts also contain elevated levels of selenium (Se), which may counteract Hg adverse effects. This study examines the fate of dietary Hg and Se in an aquaculture model fish. Biomolecular speciation analyses through hyphenated analytical approaches were conducted on the water-soluble protein fraction of key organs of juvenile rainbow trout (Oncorhynchus mykiss) exposed to various combinations of Hg and Se species, including diets containing tuna byproducts, over a six-month period. The findings shed light on the dynamics of Hg and Se compounds in fish revealing potential Hg detoxification mechanisms through complexation with Hg-biomolecules, such as cysteine, glutathione, and metallothionein. Furthermore, the trophic transfer of selenoneine is demonstrated, revealing novel opportunities for sustainable aquafeed production. Understanding the interactions between Hg and Se in aquaculture systems is crucial for optimizing feed formulations and mitigating environmental risks. This research contributes to the broader goal of advancing sustainable practices in aquaculture while addressing food security challenges.
Collapse
Affiliation(s)
- Khouloud El Hanafi
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les matériaux, Pau, France
| | - Tamara Fernández-Bautista
- Departamento de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Laurent Ouerdane
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les matériaux, Pau, France
| | - Warren T Corns
- PS Analytical, Arthur House, Crayfields Industrial Estate, Main Road, Orpington, Kent BR5 3HP, United Kingdom
| | - Maite Bueno
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les matériaux, Pau, France
| | | | - David Amouroux
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les matériaux, Pau, France
| | - Zoyne Pedrero
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Institut des Sciences Analytiques et de Physico-chimie pour l'Environnement et les matériaux, Pau, France.
| |
Collapse
|
3
|
Hei Z, Yang S, Ouyang G, Hanna J, Lepoivre M, Huynh T, Aguinaga L, Cassinat B, Maslah N, Bourge M, Golinelli-Cohen MP, Guittet O, Vallières C, Vernis L, Fenaux P, Huang ME. Targeting the redox vulnerability of acute myeloid leukaemia cells with a combination of auranofin and vitamin C. Br J Haematol 2024; 205:1017-1030. [PMID: 39087522 DOI: 10.1111/bjh.19680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by complex molecular and cytogenetic abnormalities. Pro-oxidant cellular redox status is a common hallmark of AML cells, providing a rationale for redox-based anticancer strategy. We previously discovered that auranofin (AUF), initially used for the treatment of rheumatoid arthritis and repositioned for its anticancer activity, can synergize with a pharmacological concentration of vitamin C (VC) against breast cancer cell line models. In this study, we observed that this drug combination synergistically and efficiently killed cells of leukaemic cell lines established from different myeloid subtypes. In addition to an induced elevation of reactive oxygen species and ATP depletion, a rapid dephosphorylation of 4E-BP1 and p70S6K, together with a strong inhibition of protein synthesis were early events in response to AUF/VC treatment, suggesting their implication in AUF/VC-induced cytotoxicity. Importantly, a study on 22 primary AML specimens from various AML subtypes showed that AUF/VC combinations at pharmacologically achievable concentrations were effective to eradicate primary leukaemic CD34+ cells from the majority of these samples, while being less toxic to normal cord blood CD34+ cells. Our findings indicate that targeting the redox vulnerability of AML with AUF/VC combinations could present a potential anti-AML therapeutic approach.
Collapse
Affiliation(s)
- Zhiliang Hei
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Shujun Yang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Guifang Ouyang
- Department of Hematology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jolimar Hanna
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Michel Lepoivre
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Tony Huynh
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Lorea Aguinaga
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Bruno Cassinat
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Nabih Maslah
- INSERM UMR 1131, Université Paris Cité, Hôpital Saint-Louis, IRSL, Paris, France
| | - Mickaël Bourge
- Cytometry Facility, Imagerie-Gif, Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | | | - Olivier Guittet
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Cindy Vallières
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Laurence Vernis
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Meng-Er Huang
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| |
Collapse
|
4
|
DeAngelo SL, Zhao L, Dziechciarz S, Shin M, Solanki S, Balia A, El-Derany MO, Castillo C, Qin Y, Das NK, Bell HN, Paulo JA, Zhang Y, Rossiter NJ, McCulla EC, He J, Talukder I, Ng BWL, Schafer ZT, Neamati N, Mancias JD, Koutmos M, Shah YM. Recharacterization of RSL3 reveals that the selenoproteome is a druggable target in colorectal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587381. [PMID: 38617233 PMCID: PMC11014488 DOI: 10.1101/2024.03.29.587381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Ferroptosis is a non-apoptotic form of cell death resulting from the iron-dependent accumulation of lipid peroxides. Colorectal cancer (CRC) cells accumulate high levels of intracellular iron and reactive oxygen species (ROS) and are thus particularly sensitive to ferroptosis. The compound (S)-RSL3 ([1S,3R]-RSL3) is a commonly used ferroptosis inducing compound that is currently characterized as a selective inhibitor of the selenocysteine containing enzyme (selenoprotein) Gluathione Peroxidase 4 (GPx4), an enzyme that utilizes glutathione to directly detoxify lipid peroxides. However, through chemical controls utilizing the (R) stereoisomer of RSL3 ([1R,3R]-RSL3) that does not bind GPx4, combined with inducible genetic knockdowns of GPx4 in CRC cell lines, we revealed that GPx4 dependency does not always align with (S)-RSL3 sensitivity, questioning the current characterization of GPx4 as the central regulator of ferroptosis. Utilizing affinity pull-down mass spectrometry with chemically modified (S)-RSL3 probes we discovered that the effects of (S)-RSL3 extend far beyond GPx4 inhibition, revealing that (S)-RSL3 is a broad and non-selective inhibitor of selenoproteins. To further investigate the therapeutic potential of broadly disrupting the selenoproteome as a therapeutic strategy in CRC, we employed additional chemical and genetic approaches. We found that the selenoprotein inhibitor auranofin, an FDA approved gold-salt, chemically induced oxidative cell death and ferroptosis in both in-vitro and in-vivo models of CRC. Consistent with these data, we found that AlkBH8, a tRNA-selenocysteine methyltransferase required for the translation of selenoproteins, is essential for the in-vitro growth and xenograft survival of CRC cell lines. In summary, these findings recharacterize the mechanism of action of the most commonly used ferroptosis inducing molecule, (S)-RSL3, and reveal that broad inhibition of selenoproteins is a promising novel therapeutic angle for the treatment of CRC.
Collapse
Affiliation(s)
- Stephen L. DeAngelo
- Doctoral Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Chemistry, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Liang Zhao
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Sofia Dziechciarz
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Myungsun Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States
| | - Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Andrii Balia
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
| | - Marwa O El-Derany
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cristina Castillo
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Yao Qin
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Nupur K. Das
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Hannah Noelle Bell
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States
| | - Yuezhong Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas J. Rossiter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Elizabeth C. McCulla
- Doctoral Program in Cancer Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Jianping He
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - Indrani Talukder
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Billy Wai-Lung Ng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Zachary T. Schafer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Joseph D. Mancias
- Dana-Farber Cancer Institute, Boston, Massachusetts, United States
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA. United States
| | - Markos Koutmos
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA. United States
- Department of Chemistry, University of Michigan, Ann Arbor, MI, United States
- Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
5
|
An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death Dis 2024; 15:556. [PMID: 39090114 PMCID: PMC11294602 DOI: 10.1038/s41419-024-06939-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin An
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Yu
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Li Yang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
6
|
Nakahata DH, Kanavos I, Zubiria-Ulacia M, Inague A, Salassa L, Lobinski R, Miyamoto S, Matxain JM, Ronga L, de Paiva REF. Gold-Promoted Biocompatible Selenium Arylation of Small Molecules, Peptides and Proteins. Chemistry 2024; 30:e202304050. [PMID: 38197477 DOI: 10.1002/chem.202304050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/11/2024]
Abstract
A low pKa (5.2), high polarizable volume (3.8 Å), and proneness to oxidation under ambient conditions make selenocysteine (Sec, U) a unique, natural reactive handle present in most organisms across all domains of life. Sec modification still has untapped potential for site-selective protein modification and probing. Herein we demonstrate the use of a cyclometalated gold(III) compound, [Au(bnpy)Cl2 ], in the arylation of diselenides of biological significance, with a scope covering small molecule models, peptides, and proteins using a combination of multinuclear NMR (including 77 Se NMR), and LC-MS. Diphenyl diselenide (Ph-Se)2 and selenocystine, (Sec)2 , were used for reaction optimization. This approach allowed us to demonstrate that an excess of diselenide (Au/Se-Se) and an increasing water percentage in the reaction media enhance both the conversion and kinetics of the C-Se coupling reaction, a combination that makes the reaction biocompatible. The C-Se coupling reaction was also shown to happen for the diselenide analogue of the cyclic peptide vasopressin ((Se-Se)-AVP), and the Bos taurus glutathione peroxidase (GPx1) enzyme in ammonium acetate (2 mM, pH=7.0). The reaction mechanism, studied by DFT revealed a redox-based mechanism where the C-Se coupling is enabled by the reductive elimination of the cyclometalated Au(III) species into Au(I).
Collapse
Affiliation(s)
- Douglas H Nakahata
- Donostia International Physics Center - DIPC, Paseo Manuel de Lardizabal 4, 20018, Donostia, Euskadi, Gipuzkoa, Spain
| | - Ioannis Kanavos
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux - IPREM, E2S UPPA, CNRS, Université de Pau et des Pays de l'Adour, 64053, Pau, France
| | - Maria Zubiria-Ulacia
- Donostia International Physics Center - DIPC, Paseo Manuel de Lardizabal 4, 20018, Donostia, Euskadi, Gipuzkoa, Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea Euskal Herriko Unibertsitatea UPV/EHU, Donostia, Spain, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, 20018, Donostia, Euskadi, Gipuzkoa, Spain
| | - Alex Inague
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, 05508000, SP, Brazil
| | - Luca Salassa
- Donostia International Physics Center - DIPC, Paseo Manuel de Lardizabal 4, 20018, Donostia, Euskadi, Gipuzkoa, Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea Euskal Herriko Unibertsitatea UPV/EHU, Donostia, Spain, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, 20018, Donostia, Euskadi, Gipuzkoa, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Euskadi, Bizkaia, Spain
| | - Ryszard Lobinski
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux - IPREM, E2S UPPA, CNRS, Université de Pau et des Pays de l'Adour, 64053, Pau, France
| | - Sayuri Miyamoto
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, 05508000, SP, Brazil
| | - Jon Mattin Matxain
- Donostia International Physics Center - DIPC, Paseo Manuel de Lardizabal 4, 20018, Donostia, Euskadi, Gipuzkoa, Spain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea Euskal Herriko Unibertsitatea UPV/EHU, Donostia, Spain, Euskal Herriko Unibertsitatea UPV/EHU, Paseo Manuel de Lardizabal 3, 20018, Donostia, Euskadi, Gipuzkoa, Spain
| | - Luisa Ronga
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux - IPREM, E2S UPPA, CNRS, Université de Pau et des Pays de l'Adour, 64053, Pau, France
| | - Raphael E F de Paiva
- Donostia International Physics Center - DIPC, Paseo Manuel de Lardizabal 4, 20018, Donostia, Euskadi, Gipuzkoa, Spain
| |
Collapse
|
7
|
Bramatti I, Aschner M, Branco V, Carvalho C. Exposure of human glioblastoma cells to thimerosal inhibits the thioredoxin system and decreases tumor growth-related factors. Toxicol Appl Pharmacol 2024; 484:116844. [PMID: 38325586 DOI: 10.1016/j.taap.2024.116844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Glioblastoma multiforme (GBM) is the most common, aggressive, and fatal primary malignant brain tumor in adults. The therapeutic efficacy of temozolomide (TMZ) is limited owing to frequent treatment resistance. The latter is in part related to the overexpression of redox systems such as the thioredoxin system. This system is fundamental for cell survival and proliferation, regulating hypoxia inducible factor-1alpha (HIF-1α) activity, in turn controlling vascular endothelial growth factor (VEGF), which is indispensable for tumor invasiveness, angiogenesis and microenvironment maintenance. HIF-1α can also be regulated by the signal transducer and activator of transcription 3 (STAT3), an oncogene stimulated by pro-inflammatory cytokines and growth factors. The thioredoxin system has several known inhibitors including mercury compounds such as Thimerosal (TmHg) which readily crosses the blood-brain barrier (BBB) and accumulates in the brain. Though previously used in various applications epidemiological evidence on TmHg's neurotoxicity is lacking. The objective of this study was to verify whether thimerosal is a suitable candidate for hard repurposing to control glioblastoma; therefore, the effects of this molecule were evaluated in human GBM (U87) cells. Our novel results show that TmHg decreased cellular viability (>50%) and migration (up to 90% decrease in wound closure), reduced thioredoxin reductase (TrxR/TXNRD1) and thioredoxin (Trx) activity, and increased reactive oxygen species (ROS) generation. Moreover, TmHg reduced HIF-1α expression (35%) as observed by immunofluorescence. Co-exposure of U87 cells to TmHg and TMZ reduced HIF-1α, VEGF, and phosphorylated STAT3. Consequently, TmHg alone or combined with chemotherapeutic drugs can reduce neoangiogenesis and ameliorate glioblastoma progression and treatment.
Collapse
Affiliation(s)
- Isabella Bramatti
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cristina Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
8
|
Tolbatov I, Umari P, Marrone A. Mechanism of Action of Antitumor Au(I) N-Heterocyclic Carbene Complexes: A Computational Insight on the Targeting of TrxR Selenocysteine. Int J Mol Sci 2024; 25:2625. [PMID: 38473872 DOI: 10.3390/ijms25052625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
The targeting of human thioredoxin reductase is widely recognized to be crucially involved in the anticancer properties of several metallodrugs, including Au(I) complexes. In this study, the mechanism of reaction between a set of five N-heterocyclic carbene Au(I) complexes and models of the active Sec residue in human thioredoxin reductase was investigated by means of density functional theory approaches. The study was specifically addressed to the kinetics and thermodynamics of the tiled process by aiming at elucidating and explaining the differential inhibitory potency in this set of analogous Au(I) bis-carbene complexes. While the calculated free energy profile showed a substantially similar reactivity, we found that the binding of these Au(I) bis-carbene at the active CysSec dyad in the TrxR enzyme could be subjected to steric and orientational restraints, underlining both the approach of the bis-carbene scaffold and the attack of the selenol group at the metal center. A new and detailed mechanistic insight to the anticancer activity of these Au(I) organometallic complexes was thus provided by consolidating the TrxR targeting paradigm.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Department of Physics and Astronomy, University of Padova, Via F. Marzolo 8, 35131 Padova, Italy
| | - Paolo Umari
- Department of Physics and Astronomy, University of Padova, Via F. Marzolo 8, 35131 Padova, Italy
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università "G d'Annunzio" di Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
9
|
Dos Santos HF, Paschoal DFS. S-Se oxidative addition to auranofin derivatives: a DFT study. Phys Chem Chem Phys 2024; 26:5517-5528. [PMID: 38284132 DOI: 10.1039/d3cp04913b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Oxidative addition of the S-Se bond to Au(I) complexes is discussed for a series of 26 auranofin (AF) derivatives. AF and its analogues are Au(I) complexes with recognized anticancer activity that act by binding and inhibiting the thioredoxin reductase (TrxR) enzyme. Generally, the oxidative addition to Au(I) is a sluggish reaction under mild conditions (i.e., a high activation barrier - ΔH‡), which is also verified here for AF, ΔH‡ = 33.0 kcal mol-1. However, we predicted that subtle changes in the AF ligands can make the process feasible under standard conditions. For instance, the exchange of -PEt3 by -P(Et2)(OEt), which is a weaker electron σ-donor, reduced the activation barrier to 17.1 kcal mol-1. Furthermore, substitution of the -SAtg ligand by -Cl- leads to a ΔH‡ value of 22.5 kcal mol-1. Overall, the reaction is driven by the nucleophilic attack of the S-Se bond on the Au(I) center, attributed mainly to the charge transfer (4p)Se → (6p)Au, which characterizes the addition step. At the transition state (TS) point, the (5d)Au → σ*(S-Se) charge transfer becomes relevant, facilitating the S-Se bond breakage and the oxidation step. In addition to the electron transfers, the strain energy to deform the linear Au(I) geometry to the tetracoordinated Au(III) arrangement in the TS structure plays a primary role in explaining the trends in the activation barriers. Finally, the activation barrier (ΔH‡) and reaction energy (ΔH°) were correlated for most of the complexes studied, which suggests that the reaction passes through a late or product-like TS and, therefore, the steric and electronic factors affecting ΔH‡ also act on ΔH°. Overall, the results presented here might open up a new field of investigation for interactions between AF derivatives and TrxR, which contributes to a full understanding of the biological mechanism of action of these species.
Collapse
Affiliation(s)
- Hélio F Dos Santos
- NEQC: Núcleo de Estudos em Química Computacional, Departamento de Química - ICE, Universidade Federal de Juiz de Fora, Campus Universitário, 36.036-900, Juiz de Fora, MG, Brazil.
| | - Diego F S Paschoal
- NQTCM: Núcleo de Química Teórica e Computacional de Macaé, Polo Ajuda, Instituto Multidisciplinar de Química, Centro Multidisciplinar UFRJ-Macaé, Universidade Federal do Rio de Janeiro, 27.971-525, Macaé, RJ, Brazil
| |
Collapse
|
10
|
Barbosa NV, Aschner M, Tinkov AA, Farina M, da Rocha JBT. Should ebselen be considered for the treatment of mercury intoxication? A minireview. Toxicol Mech Methods 2024; 34:1-12. [PMID: 37731353 PMCID: PMC10841883 DOI: 10.1080/15376516.2023.2258958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023]
Abstract
Mercury is a ubiquitous environmental contaminant and can be found in inorganic (Hg0, Hg+ and Hg2+) and organic forms (chiefly CH3Hg+ or MeHg+). The main route of human, mammals and bird exposure occurs via predatory fish ingestion. Occupational exposure to Hg0 (and Hg2+) can also occur; furthermore, in gold mining areas the exposure to inorganic Hg can also be high. The toxicity of electrophilic forms of Hg (E+Hg) is mediated by disruption of thiol (-SH)- or selenol (-SeH)-containing proteins. The therapeutic approaches to treat methylmercury (MeHg+), Hg0 and Hg2+ are limited. Here we discuss the potential use of ebselen as a potential therapeutic agent to lower the body burden of Hg in man. Ebselen is a safe drug for humans and has been tested in clinical trials (for instance, brain ischemia, noise-induce hearing loss, diabetes complications, bipolar disorders) at doses varying from 400 to 3600 mg per day. Two clinical trials with ebselen in moderate and severe COVID are also approved. Ebselen can be metabolized to an intermediate with -SeH (selenol) functional group, which has a greater affinity to electrophilic Hg (E+Hg) forms than the available thiol-containing therapeutic agents. Accordingly, as observed in vitro and rodent models in vivo, Ebselen exhibited protective effects against MeHg+, indicating its potential as a therapeutic agent to treat MeHg+ overexposure. The combined use of ebselen with thiol-containing molecules (e.g. N-acetylcysteine and enaramide)) is also commented, because they can have synergistic protective effects against MeHg+.
Collapse
Affiliation(s)
- Nilda V. Barbosa
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexey A. Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - João Batista Teixeira da Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
- Departamento de Bioquímica, Instituto Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
11
|
James AK, Popescu BF, Weng M, Myers GJ, O'Donoghue JL, Watson GE, Pickering IJ, George GN. Synchrotron X-ray methods in the study of mercury neurotoxicology. Neurotoxicology 2023; 99:129-138. [PMID: 37802190 DOI: 10.1016/j.neuro.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/22/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023]
Abstract
In situ methods are valuable in all fields of research. In toxicology, the importance of dose is well known, elevating the need for in situ techniques to measure levels of toxicants and their byproducts in precise anatomically identifiable locations. More recently, additional emphasis has been placed on the value of techniques which can detect chemical form or speciation, which is equally important in the toxicology of a chemical compound. Many important but conventional methods risk losing valuable information due to extractions, digestions, or the general reliance on mobile phases. Few analytical tools possess the power and diversity of X-ray methods as in-situ methods. Here we present an overview, intended for toxicologists and pathologists, of a variety of synchrotron X-ray methods for determining in situ chemical form and distribution of heavier elements. The versatility and range of these synchrotron techniques, which are both established and emerging, is demonstrated in the context of the study of neurotoxicology of mercury, a global pollutant with the ability to harm both human health and the environment.
Collapse
Affiliation(s)
- Ashley K James
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon City Hospital, Saskatoon, SK S7K 0M7, Canada; Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, SK S7N 5E2, Canada.
| | - Bogdan F Popescu
- Cameco MS Neuroscience Research Centre, University of Saskatchewan, Saskatoon City Hospital, Saskatoon, SK S7K 0M7, Canada
| | - Monica Weng
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Gary J Myers
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA; Departments of Neurology and Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - John L O'Donoghue
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Gene E Watson
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA; Eastman Institute for Oral Health, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Ingrid J Pickering
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada; Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada
| | - Graham N George
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada; Department of Chemistry, University of Saskatchewan, Saskatoon, SK S7N 5C9, Canada.
| |
Collapse
|
12
|
Andor A, Mohanraj M, Pató ZA, Úri K, Biri-Kovács B, Cheng Q, Arnér ESJ. TXNL1 has dual functions as a redox active thioredoxin-like protein as well as an ATP- and redox-independent chaperone. Redox Biol 2023; 67:102897. [PMID: 37804695 PMCID: PMC10570131 DOI: 10.1016/j.redox.2023.102897] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 10/09/2023] Open
Abstract
TXNL1 (also named TRP32, for thioredoxin related protein of 32 kDa) is a cytosolic thioredoxin-fold protein expressed in all cell types and conserved from yeast to mammals, but with yet poorly known function. Here, we expressed and purified human TXNL1 together with several Cys-to-Ser variants, characterizing their enzymatic properties. TXNL1 could reduce disulfides in insulin, cystine and glutathione disulfide (GSSG) in reactions coupled to thioredoxin reductase (TXNRD1, TrxR1) using NADPH, similarly to thioredoxin (TXN, Trx1), but with lower catalytic efficacy due to at least one order of magnitude higher Km of TrxR1 for TXNL1 compared to Trx1. However, in sharp contrast to Trx1, we found that TXNL1 also had efficient chaperone activity that did not require ATP. TXNL1 made non-covalent complexes with reduced insulin, thereby keeping it in solution, and TXNL1 provided chaperone function towards whole cell lysate proteins by preventing their aggregation during heating. The chaperone activities of TXNL1 did not require its redox activity or any dithiol-disulfide exchange reactions, as revealed using Cys-to-Ser substituted variants, as well as a maintained chaperone activity of TXNL1 also in the absence of TrxR1 and NADPH. These results reveal that TXNL1 has dual functions, supporting TrxR1-driven redox activities in disulfide reduction reactions, as well as being an ATP-independent chaperone that does not require involvement of its redox activity.
Collapse
Affiliation(s)
- Attila Andor
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Mahendravarman Mohanraj
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Zsuzsanna Anna Pató
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Katalin Úri
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Beáta Biri-Kovács
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Elias S J Arnér
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Division of Biochemistry, Department of Medical Biochemistry, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
13
|
Müller VVL, Simpson PV, Peng K, Basu U, Moreth D, Nagel C, Türck S, Oehninger L, Ott I, Schatzschneider U. Taming the Biological Activity of Pd(II) and Pt(II) Complexes with Triazolato "Protective" Groups: 1H, 77Se Nuclear Magnetic Resonance and X-ray Crystallographic Model Studies with Selenocysteine to Elucidate Differential Thioredoxin Reductase Inhibition. Inorg Chem 2023; 62:16203-16214. [PMID: 37713601 DOI: 10.1021/acs.inorgchem.3c02701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
The biological activity of Pd(II) and Pt(II) complexes toward three different cancer cell lines as well as inhibition of selenoenzyme thioredoxin reductase (TrxR) was modulated in an unexpected way by the introduction of triazolate as a "protective group" to the inner metal coordination sphere using the iClick reaction of [M(N3)(terpy)]PF6 [M = Pd(II) or Pt(II) and terpy = 2,2':6',2″-terpyridine] with an electron-poor alkyne. In a cell proliferation assay using A549, HT-29, and MDA-MB-231 human cancer cell lines, the palladium compound was significantly more potent than the isostructural platinum analogue and exhibited submicromolar activity on the most responsive cell line. This difference was also reflected in the inhibitory efficiency toward TrxR with IC50 values of 0.1 versus 5.4 μM for the Pd(II) and Pt(II) complexes, respectively. UV/Vis kinetic studies revealed that the Pt compound binds to selenocysteine faster than to cysteine [k = (22.9 ± 0.2)·10-3 vs (7.1 ± 0.2)·10-3 s-1]. Selective triazolato ligand exchange of the title compounds with cysteine (Hcys) and selenocysteine (Hsec)─but not histidine (His) and 9-ethylguanine (9EtG)─was confirmed by 1H, 77Se, and 195Pt NMR spectroscopy. Crystal structures of three of the four ligand exchange products were obtained, including [Pt(sec)(terpy)]PF6 as the first metal complex of selenocysteine to be structurally characterized.
Collapse
Affiliation(s)
- Victoria V L Müller
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Peter V Simpson
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Kun Peng
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Uttara Basu
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany
| | - Dominik Moreth
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Christoph Nagel
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Sebastian Türck
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany
| | - Luciano Oehninger
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany
| | - Ingo Ott
- Institut für Medizinische und Pharmazeutische Chemie, Technische Universität Braunschweig, Beethovenstr. 55, D-38106 Braunschweig, Germany
| | - Ulrich Schatzschneider
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|
14
|
Liu M, Sun S, Meng Y, Wang L, Liu H, Shi W, Zhang Q, Xu W, Sun B, Xu J. Benzophenanthridine Alkaloid Chelerythrine Elicits Necroptosis of Gastric Cancer Cells via Selective Conjugation at the Redox Hyperreactive C-Terminal Sec 498 Residue of Cytosolic Selenoprotein Thioredoxin Reductase. Molecules 2023; 28:6842. [PMID: 37836684 PMCID: PMC10574601 DOI: 10.3390/molecules28196842] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/23/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
Targeting thioredoxin reductase (TXNRD) with low-weight molecules is emerging as a high-efficacy anti-cancer strategy in chemotherapy. Sanguinarine has been reported to inhibit the activity of TXNRD1, indicating that benzophenanthridine alkaloid is a fascinating chemical entity in the field of TXNRD1 inhibitors. In this study, the inhibition of three benzophenanthridine alkaloids, including chelerythrine, sanguinarine, and nitidine, on recombinant TXNRD1 was investigated, and their anti-cancer mechanisms were revealed using three gastric cancer cell lines. Chelerythrine and sanguinarine are more potent inhibitors of TXNRD1 than nitidine, and the inhibitory effects take place in a dose- and time-dependent manner. Site-directed mutagenesis of TXNRD1 and in vitro inhibition analysis proved that chelerythrine or sanguinarine is primarily bound to the Sec498 residue of the enzyme, but the neighboring Cys497 and remaining N-terminal redox-active cysteines could also be modified after the conjugation of Sec498. With high similarity to sanguinarine, chelerythrine exhibited cytotoxic effects on multiple gastric cancer cell lines and suppressed the proliferation of tumor spheroids derived from NCI-N87 cells. Chelerythrine elevated cellular levels of reactive oxygen species (ROS) and induced endoplasmic reticulum (ER) stress. Moreover, the ROS induced by chelerythrine could be completely suppressed by the addition of N-acetyl-L-cysteine (NAC), and the same is true for sanguinarine. Notably, Nec-1, an RIPK1 inhibitor, rescued the chelerythrine-induced rapid cell death, indicating that chelerythrine triggers necroptosis in gastric cancer cells. Taken together, this study demonstrates that chelerythrine is a novel inhibitor of TXNRD1 by targeting Sec498 and possessing high anti-tumor properties on multiple gastric cancer cell lines by eliciting necroptosis.
Collapse
Affiliation(s)
- Minghui Liu
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Shibo Sun
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Yao Meng
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Ling Wang
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Haowen Liu
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Wuyang Shi
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Qiuyu Zhang
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| | - Weiping Xu
- School of Ocean Science and Technology (OST), Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian University of Technology, Panjin 124221, China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering (CE), Dalian University of Technology, Dalian 116023, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences (LPS), Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin 124221, China
| |
Collapse
|
15
|
Bernabeu de Maria M, Tesauro D, Prencipe F, Saviano M, Messori L, Enjalbal C, Lobinski R, Ronga L. Disclosing the Preferential Mercury Chelation by SeCys Containing Peptides over Their Cys Analogues. Inorg Chem 2023; 62:14980-14990. [PMID: 37651565 DOI: 10.1021/acs.inorgchem.3c01708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Methylmercury, mercury (II), and mercury (I) chlorides were found to react with vasopressin, a nonapeptide hormone cyclized by two cysteine residues, and its mono- and diselenium analogues to form several mercury-peptide adducts. The replacement of Cys by SeCys in vasopressin increased the reactivity toward methylmercury, with the predominant formation of -Se/S-Hg-Se-bridged structures and the consequent demethylation of methylmercury. In competitive experiments, CH3HgCl reacted preferentially with the diselenium analogue rather than with vasopressin. The diselenium peptide also showed the capability to displace the CH3Hg moiety bound to S in vasopressin. These results open a promising perspective for the use of selenopeptides for methylmercury chelation and detoxification strategies.
Collapse
Affiliation(s)
| | - Diego Tesauro
- Department of Pharmacy and CIRPeB, Università Degli Studi di Napoli Federico II, 49 80131 Naples, Italy
| | | | | | - Luigi Messori
- Department of Chemistry, Università Degli Studi di Firenze, 50019 Sesto Fiorentino, Italy
| | - Christine Enjalbal
- IBMM, Université de Montpellier, CNRS, ENSCM, UMR 5247, 34293 Montpellier Cedex 5, France
| | - Ryszard Lobinski
- Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64000 Pau, France
- Warsaw University of Technology, 00-664 Warsaw, Poland
| | - Luisa Ronga
- Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64000 Pau, France
| |
Collapse
|
16
|
Ratia C, Ballén V, Gabasa Y, Soengas RG, Velasco-de Andrés M, Iglesias MJ, Cheng Q, Lozano F, Arnér ESJ, López-Ortiz F, Soto SM. Novel gold(III)-dithiocarbamate complex targeting bacterial thioredoxin reductase: antimicrobial activity, synergy, toxicity, and mechanistic insights. Front Microbiol 2023; 14:1198473. [PMID: 37333656 PMCID: PMC10272563 DOI: 10.3389/fmicb.2023.1198473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Antimicrobial resistance is a pressing global concern that has led to the search for new antibacterial agents with novel targets or non-traditional approaches. Recently, organogold compounds have emerged as a promising class of antibacterial agents. In this study, we present and characterize a (C^S)-cyclometallated Au(III) dithiocarbamate complex as a potential drug candidate. Methods and results The Au(III) complex was found to be stable in the presence of effective biological reductants, and showed potent antibacterial and antibiofilm activity against a wide range of multidrug-resistant strains, particularly gram-positive strains, and gram-negative strains when used in combination with a permeabilizing antibiotic. No resistant mutants were detected after exposing bacterial cultures to strong selective pressure, indicating that the complex may have a low propensity for resistance development. Mechanistic studies indicate that the Au(III) complex exerts its antibacterial activity through a multimodal mechanism of action. Ultrastructural membrane damage and rapid bacterial uptake suggest direct interactions with the bacterial membrane, while transcriptomic analysis identified altered pathways related to energy metabolism and membrane stability including enzymes of the TCA cycle and fatty acid biosynthesis. Enzymatic studies further revealed a strong reversible inhibition of the bacterial thioredoxin reductase. Importantly, the Au(III) complex demonstrated low cytotoxicity at therapeutic concentrations in mammalian cell lines, and showed no acute in vivo toxicity in mice at the doses tested, with no signs of organ toxicity. Discussion Overall, these findings highlight the potential of the Au(III)-dithiocarbamate scaffold as a basis for developing novel antimicrobial agents, given its potent antibacterial activity, synergy, redox stability, inability to produce resistant mutants, low toxicity to mammalian cells both in vitro and in vivo, and non-conventional mechanism of action.
Collapse
Affiliation(s)
- Carlos Ratia
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Victoria Ballén
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Yaiza Gabasa
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
| | - Raquel G. Soengas
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | | | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Lozano
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Servei d’Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain
- Department de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Elias S. J. Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Selenoprotein Research and the National Tumor Biology Laboratory, Budapest, Hungary
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, Almería, Spain
| | - Sara M. Soto
- Barcelona Institute for Global Health (ISGlobal), Universitat de Barcelona, Barcelona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
17
|
Mertens RT, Gukathasan S, Arojojoye AS, Olelewe C, Awuah SG. Next Generation Gold Drugs and Probes: Chemistry and Biomedical Applications. Chem Rev 2023; 123:6612-6667. [PMID: 37071737 PMCID: PMC10317554 DOI: 10.1021/acs.chemrev.2c00649] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
The gold drugs, gold sodium thiomalate (Myocrisin), aurothioglucose (Solganal), and the orally administered auranofin (Ridaura), are utilized in modern medicine for the treatment of inflammatory arthritis including rheumatoid and juvenile arthritis; however, new gold agents have been slow to enter the clinic. Repurposing of auranofin in different disease indications such as cancer, parasitic, and microbial infections in the clinic has provided impetus for the development of new gold complexes for biomedical applications based on unique mechanistic insights differentiated from auranofin. Various chemical methods for the preparation of physiologically stable gold complexes and associated mechanisms have been explored in biomedicine such as therapeutics or chemical probes. In this Review, we discuss the chemistry of next generation gold drugs, which encompasses oxidation states, geometry, ligands, coordination, and organometallic compounds for infectious diseases, cancer, inflammation, and as tools for chemical biology via gold-protein interactions. We will focus on the development of gold agents in biomedicine within the past decade. The Review provides readers with an accessible overview of the utility, development, and mechanism of action of gold-based small molecules to establish context and basis for the thriving resurgence of gold in medicine.
Collapse
Affiliation(s)
- R Tyler Mertens
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Sailajah Gukathasan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Adedamola S Arojojoye
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Chibuzor Olelewe
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Markey Cancer Center, Lexington, Kentucky 40536, United States
| |
Collapse
|
18
|
Cheff DM, Cheng Q, Guo H, Travers J, Klumpp-Thomas C, Shen M, Arnér ESJ, Hall MD. Development of an assay pipeline for the discovery of novel small molecule inhibitors of human glutathione peroxidases GPX1 and GPX4. Redox Biol 2023; 63:102719. [PMID: 37244126 DOI: 10.1016/j.redox.2023.102719] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/29/2023] Open
Abstract
Selenoprotein glutathione peroxidases (GPX), like ubiquitously expressed GPX1 and the ferroptosis modulator GPX4, enact antioxidant activities by reducing hydroperoxides using glutathione. Overexpression of these enzymes is common in cancer and can be associated with the development of resistance to chemotherapy. GPX1 and GPX4 inhibitors have thus shown promise as anti-cancer agents, and targeting other GPX isoforms may prove equally beneficial. Existing inhibitors are often promiscuous, or modulate GPXs only indirectly, so novel direct inhibitors identified through screening against GPX1 and GPX4 could be valuable. Here, we developed optimized glutathione reductase (GR)-coupled GPX assays for the biochemical high-throughput screen (HTS) of almost 12,000 compounds with proposed mechanisms of action. Initial hits were triaged using a GR counter-screen, assessed for isoform specificity against an additional GPX isoform, GPX2, and were assessed for general selenocysteine-targeting activity using a thioredoxin reductase (TXNRD1) assay. Importantly, 70% of the GPX1 inhibitors identified in the primary screen, including several cephalosporin antibiotics, were found to also inhibit TXNRD1, while auranofin, previously known as a TXNRD1 inhibitor, also inhibited GPX1 (but not GPX4). Additionally, every GPX1 inhibitor identified (including omapatrilat, tenatoprazole, cefoxitin and ceftibuten) showed similar inhibitory activity against GPX2. Some compounds inhibiting GPX4 but not GPX1 or GPX2, also inhibited TXNRD1 (26%). Compounds only inhibiting GPX4 included pranlukast sodium hydrate, lusutrombopag, brilanestrant, simeprevir, grazoprevir (MK-5172), paritaprevir, navitoclax, venetoclax and VU0661013. Two compounds (metamizole sodium and isoniazid sodium methanesulfate) inhibited all three GPXs but not TXNRD1, while 2,3-dimercaptopropanesulfonate, PI4KIII beta inhibitor 3, SCE-2174 and cefotetan sodium inhibited all tested selenoproteins (but not GR). The detected overlaps in chemical space suggest that the counter screens introduced here should be imperative for identification of specific GPX inhibitors. With this approach, we could indeed identify novel GPX1/GPX2- or GPX4-specific inhibitors, thus presenting a validated pipeline for future identification of specific selenoprotein-targeting agents. Our study also identified GPX1/GPX2, GPX4 and/or TXNRD1 as targets for several previously developed pharmacologically active compounds.
Collapse
Affiliation(s)
- Dorian M Cheff
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States; Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden
| | - Hui Guo
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Jameson Travers
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Carleen Klumpp-Thomas
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Min Shen
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE, 171 77, Stockholm, Sweden; Department of Selenoprotein Research and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary.
| | - Matthew D Hall
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institute of Health, 9800 Medical Center Drive, Rockville, MD, 20850, United States.
| |
Collapse
|
19
|
Tolbatov I, Marrone A. Auranofin Targeting the NDM-1 Beta-Lactamase: Computational Insights into the Electronic Configuration and Quasi-Tetrahedral Coordination of Gold Ions. Pharmaceutics 2023; 15:pharmaceutics15030985. [PMID: 36986846 PMCID: PMC10057648 DOI: 10.3390/pharmaceutics15030985] [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: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Recently, the well-characterized metallodrug auranofin has been demonstrated to restore the penicillin and cephalosporin sensitivity in resistant bacterial strains via the inhibition of the NDM-1 beta-lactamase, which is operated via the Zn/Au substitution in its bimetallic core. The resulting unusual tetrahedral coordination of the two ions was investigated via the density functional theory calculations. By assessing several charge and multiplicity schemes, coupled with on/off constraining the positions of the coordinating residues, it was demonstrated that the experimental X-ray structure of the gold-bound NDM-1 is consistent with either Au(I)-Au(I) or Au(II)-Au(II) bimetallic moieties. The presented results suggest that the most probable mechanism for the auranofin-based Zn/Au exchange in NDM-1 includes the early formation of the Au(I)-Au(I) system, superseded by oxidation yielding the Au(II)-Au(II) species bearing the highest resemblance to the X-ray structure.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Av. Paisos Catalans 16, 43007 Tarragona, Spain
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università degli Studi "G. D'Annunzio" Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
20
|
Yang H, Wang H, Feng J, Liao J, Lu Y. Discovery of novel inhibition site centered on 114-bit tryptophan of Thioredoxin reductase 1 through computer-aided drug design. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
21
|
Selenol (-SeH) as a target for mercury and gold in biological systems: Contributions of mass spectrometry and atomic spectroscopy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
22
|
Gencheva R, Cheng Q, Arnér ESJ. Thioredoxin reductase selenoproteins from different organisms as potential drug targets for treatment of human diseases. Free Radic Biol Med 2022; 190:320-338. [PMID: 35987423 DOI: 10.1016/j.freeradbiomed.2022.07.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/25/2022] [Accepted: 07/26/2022] [Indexed: 11/15/2022]
Abstract
Human thioredoxin reductase (TrxR) is a selenoprotein with a central role in cellular redox homeostasis, utilizing a highly reactive and solvent-exposed selenocysteine (Sec) residue in its active site. Pharmacological modulation of TrxR can be obtained with several classes of small compounds showing different mechanisms of action, but most often dependent upon interactions with its Sec residue. The clinical implications of TrxR modulation as mediated by small compounds have been studied in diverse diseases, from rheumatoid arthritis and ischemia to cancer and parasitic infections. The possible involvement of TrxR in these diseases was in some cases serendipitously discovered, by finding that existing clinically used drugs are also TrxR inhibitors. Inhibiting isoforms of human TrxR is, however, not the only strategy for human disease treatment, as some pathogenic parasites also depend upon Sec-containing TrxR variants, including S. mansoni, B. malayi or O. volvulus. Inhibiting parasite TrxR has been shown to selectively kill parasites and can thus become a promising treatment strategy, especially in the context of quickly emerging resistance towards other drugs. Here we have summarized the basis for the targeting of selenoprotein TrxR variants with small molecules for therapeutic purposes in different human disease contexts. We discuss how Sec engagement appears to be an indispensable part of treatment efficacy and how some therapeutically promising compounds have been evaluated in preclinical or clinical studies. Several research questions remain before a wider application of selenoprotein TrxR inhibition as a first-line treatment strategy might be developed. These include further mechanistic studies of downstream effects that may mediate treatment efficacy, identification of isoform-specific enzyme inhibition patterns for some given therapeutic compounds, and the further elucidation of cell-specific effects in disease contexts such as in the tumor microenvironment or in host-parasite interactions, and which of these effects may be dependent upon the specific targeting of Sec in distinct TrxR isoforms.
Collapse
Affiliation(s)
- Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary.
| |
Collapse
|
23
|
Banerjee M, Chakravarty D, Kalwani P, Ballal A. Voyage of selenium from environment to life: Beneficial or toxic? J Biochem Mol Toxicol 2022; 36:e23195. [PMID: 35976011 DOI: 10.1002/jbt.23195] [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: 04/21/2022] [Revised: 06/22/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
Selenium (Se), a naturally occurring metalloid, is an essential micronutrient for life as it is incorporated as selenocysteine in proteins. Although beneficial at low doses, Se is hazardous at high concentrations and poses a serious threat to various ecosystems. Due to this contrasting 'dual' nature, Se has garnered the attention of researchers wishing to unravel its puzzling properties. In this review, we describe the impact of selenium's journey from environment to diverse biological systems, with an emphasis on its chemical advantage. We describe the uneven distribution of Se and how this affects the bioavailability of this element, which, in turn, profoundly affects the habitat of a region. Once taken up, the subsequent incorporation of Se into proteins as selenocysteine and its antioxidant functions are detailed here. The causes of improved protein function due to the incorporation of redox-active Se atom (instead of S) are examined. Subsequently, the reasons for the deleterious effects of Se, which depend on its chemical form (organo-selenium or the inorganic forms) in different organisms are elaborated. Although Se is vital for the function of many antioxidant enzymes, how the pro-oxidant nature of Se can be potentially exploited in different therapies is highlighted. Furthermore, we succinctly explain how the presence of Se in biological systems offsets the toxic effects of heavy metal mercury. Finally, the different avenues of research that are fundamental to expand our understanding of selenium biology are suggested.
Collapse
Affiliation(s)
- Manisha Banerjee
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Dhiman Chakravarty
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Prakash Kalwani
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Anand Ballal
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
24
|
James AK, Dolgova NV, Nehzati S, Korbas M, Cotelesage JJH, Sokaras D, Kroll T, O’Donoghue JL, Watson GE, Myers GJ, Pickering IJ, George GN. Molecular Fates of Organometallic Mercury in Human Brain. ACS Chem Neurosci 2022; 13:1756-1768. [PMID: 35543423 PMCID: PMC9977140 DOI: 10.1021/acschemneuro.2c00166] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mercury is ubiquitous in the environment, with rising levels due to pollution and climate change being a current global concern. Many mercury compounds are notorious for their toxicity, with the potential of organometallic mercury compounds for devastating effects on the structures and functions of the central nervous system being of particular concern. Chronic exposure of human populations to low levels of methylmercury compounds occurs through consumption of fish and other seafood, although the health consequences, if any, from this exposure remain controversial. We have used high energy resolution fluorescence detected X-ray absorption spectroscopy to determine the speciation of mercury and selenium in human brain tissue. We show that the molecular fate of mercury differs dramatically between individuals who suffered acute organometallic mercury exposure (poisoning) and individuals with chronic low-level exposure from a diet rich in marine fish. For long-term low-level methylmercury exposure from fish consumption, mercury speciation in brain tissue shows methylmercury coordinated to an aliphatic thiolate, resembling the coordination environment observed in marine fish. In marked contrast, for short-term high-level exposure, we observe the presence of biologically less available mercuric selenide deposits, confirmed by X-ray fluorescence imaging, as well as mercury(II)-bis-thiolate complexes, which may be signatures of severe poisoning in humans. These differences between low-level and high-level exposures challenge the relevance of studies involving acute exposure as a proxy for low-level chronic exposure.
Collapse
Affiliation(s)
- Ashley K. James
- Toxicology Centre, 44 Campus Drive, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Natalia V. Dolgova
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Susan Nehzati
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Malgorzata Korbas
- Canadian Light Source, 44 Innovation Blvd, Saskatoon, Saskatchewan S7N 2V3, Canada
| | - Julien J. H. Cotelesage
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, USA
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, USA
| | - John L. O’Donoghue
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
| | - Gene E. Watson
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Eastman Institute for Oral Health, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
| | - Gary J. Myers
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Departments of Neurology and Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
| | - Ingrid J. Pickering
- Toxicology Centre, 44 Campus Drive, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N. George
- Toxicology Centre, 44 Campus Drive, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
25
|
Fata F, Gencheva R, Cheng Q, Lullo R, Ardini M, Silvestri I, Gabriele F, Ippoliti R, Bulman CA, Sakanari JA, Williams DL, Arnér ESJ, Angelucci F. Biochemical and structural characterizations of thioredoxin reductase selenoproteins of the parasitic filarial nematodes Brugia malayi and Onchocerca volvulus. Redox Biol 2022; 51:102278. [PMID: 35276442 PMCID: PMC8914392 DOI: 10.1016/j.redox.2022.102278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/02/2022] [Indexed: 01/21/2023] Open
Abstract
Enzymes in the thiol redox systems of microbial pathogens are promising targets for drug development. In this study we characterized the thioredoxin reductase (TrxR) selenoproteins from Brugia malayi and Onchocerca volvulus, filarial nematode parasites and causative agents of lymphatic filariasis and onchocerciasis, respectively. The two filarial enzymes showed similar turnover numbers and affinities for different thioredoxin (Trx) proteins, but with a clear preference for the autologous Trx. Human TrxR1 (hTrxR1) had a high and similar specific activity versus the human and filarial Trxs, suggesting that, in vivo, hTrxR1 could possibly be the reducing agent of parasite Trxs once they are released into the host. Both filarial TrxRs were efficiently inhibited by auranofin and by a recently described inhibitor of human TrxR1 (TRi-1), but not as efficiently by the alternative compound TRi-2. The enzyme from B. malayi was structurally characterized also in complex with NADPH and auranofin, producing the first crystallographic structure of a nematode TrxR. The protein represents an unusual fusion of a mammalian-type TrxR protein architecture with an N-terminal glutaredoxin-like (Grx) domain lacking typical Grx motifs. Unlike thioredoxin glutathione reductases (TGRs) found in platyhelminths and mammals, which are also Grx-TrxR domain fusion proteins, the TrxRs from the filarial nematodes lacked glutathione disulfide reductase and Grx activities. The structural determinations revealed that the Grx domain of TrxR from B. malayi contains a cysteine (C22), conserved in TrxRs from clade IIIc nematodes, that directly interacts with the C-terminal cysteine-selenocysteine motif of the homo-dimeric subunit. Interestingly, despite this finding we found that altering C22 by mutation to serine did not affect enzyme catalysis. Thus, although the function of the Grx domain in these filarial TrxRs remains to be determined, the results obtained provide insights on key properties of this important family of selenoprotein flavoenzymes that are potential drug targets for treatment of filariasis.
Collapse
Affiliation(s)
- Francesca Fata
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Rachel Lullo
- Dept. of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Matteo Ardini
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Ilaria Silvestri
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Federica Gabriele
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Rodolfo Ippoliti
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy
| | - Christina A Bulman
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Judy A Sakanari
- Dept. of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - David L Williams
- Dept. of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Institute of Oncology, 1122, Budapest, Hungary
| | - Francesco Angelucci
- Dept. of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, 67100, Italy.
| |
Collapse
|
26
|
Nehzati S, Dolgova NV, Young CG, James AK, Cotelesage JJH, Sokaras D, Kroll T, Qureshi M, Pickering IJ, George GN. Mercury Lα1 High Energy Resolution Fluorescence Detected X-ray Absorption Spectroscopy: A Versatile Speciation Probe for Mercury. Inorg Chem 2022; 61:5201-5214. [PMID: 35073478 PMCID: PMC9962031 DOI: 10.1021/acs.inorgchem.1c03196] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mercury is in some sense an enigmatic element. The element and some of its compounds are a natural part of the biogeochemical cycle; while many of these can be deadly poisons at higher levels, environmental levels in the absence of anthropogenic contributions would generally be below the threshold for concern. However, mercury pollution, particularly from burning fossil fuels such as coal, is providing dramatic and increasing emissions into the environment. Because of this, the environmental chemistry and toxicology of mercury are of growing importance, with the fate of mercury being vitally dependent upon its speciation. X-ray absorption spectroscopy (XAS) provides a powerful tool for in situ chemical speciation, but is severely limited by poor spectroscopic energy resolution. Here, we provide a systematic examination of mercury Lα1 high energy resolution fluorescence detected XAS (HERFD-XAS) as an approach for chemical speciation of mercury, in quantitative comparison with conventional Hg LIII-edge XAS. We show that, unlike some lighter elements, chemical shifts in the Lα1 X-ray fluorescence energy can be safely neglected, so that mercury Lα1 HERFD-XAS can be treated simply as a high-resolution version of conventional XAS. We present spectra of a range of mercury compounds that may be relevant to the environmental and life science research and show that density functional theory can produce adequate simulations of the spectra. We discuss strengths and limitations of the method and quantitatively demonstrate improvements both in speciation for complex mixtures and in background rejection for low concentrations.
Collapse
Affiliation(s)
- Susan Nehzati
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Present Address: MAX IV Laboratory, Lund University, Fotongatan 2, 221 00 Lund, Sweden
| | - Natalia V. Dolgova
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Present Address: Calibr - California Institute for Biomedical Research, Scripps Research, La Jolla, California 92037, USA
| | - Charles G. Young
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Ashley K. James
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Julien J. H. Cotelesage
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, USA
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, USA
| | - Muhammad Qureshi
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, USA
| | - Ingrid J. Pickering
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N. George
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
27
|
Kalimuthu K, Keerthana CK, Mohan M, Arivalagan J, Christyraj JRSS, Firer MA, Choudry MHA, Anto RJ, Lee YJ. The emerging role of selenium metabolic pathways in cancer: New therapeutic targets for cancer. J Cell Biochem 2022; 123:532-542. [PMID: 34935169 PMCID: PMC8940641 DOI: 10.1002/jcb.30196] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 01/10/2023]
Abstract
Selenium (Se) is incorporated into the body via the selenocysteine (Sec) biosynthesis pathway, which is critical in the synthesis of selenoproteins, such as glutathione peroxidases and thioredoxin reductases. Selenoproteins, which play a key role in several biological processes, including ferroptosis, drug resistance, endoplasmic reticulum stress, and epigenetic processes, are guided by Se uptake. In this review, we critically analyze the molecular mechanisms of Se metabolism and its potential as a therapeutic target for cancer. Sec insertion sequence binding protein 2 (SECISBP2), which is a positive regulator for the expression of selenoproteins, would be a novel prognostic predictor and an alternate target for cancer. We highlight strategies that attempt to develop a novel Se metabolism-based approach to uncover a new metabolic drug target for cancer therapy. Moreover, we expect extensive clinical use of SECISBP2 as a specific biomarker in cancer therapy in the near future. Of note, scientists face additional challenges in conducting successful research, including investigations on anticancer peptides to target SECISBP2 intracellular protein.
Collapse
Affiliation(s)
- Kalishwaralal Kalimuthu
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | | | - Manikandan Mohan
- College of Pharmacy, University of Georgia, Athens, GA, USA
- VAXIGEN International Research Center Private Limited, INDIA
| | - Jaison Arivalagan
- Department of Chemistry, Molecular Biosciences and Proteomics Center of Excellence, Northwestern University, Evanston, IL, 60208, USA
| | - Johnson Retnaraj Samuel Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamilnadu, India
| | - Michael A Firer
- Dept. Chemical Engineering, Ariel University, 40700, Ariel, Israel
- Adelson School of Medicine, Ariel University, Ariel, 40700, Israel
- Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel
| | - M. Haroon A Choudry
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Yong J Lee
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Tolbatov I, Marrone A. Selenocysteine of thioredoxin reductase as the primary target for the antitumor metallodrugs: A computational point of view. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Expanding the armory for treating lymphoma: Targeting redox cellular status through thioredoxin reductase inhibition. Pharmacol Res 2022; 177:106134. [DOI: 10.1016/j.phrs.2022.106134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022]
|
30
|
Pan J, Li X, Wei Y, Ni L, Xu B, Deng Y, Yang T, Liu W. Advances on the Influence of Methylmercury Exposure during Neurodevelopment. Chem Res Toxicol 2022; 35:43-58. [PMID: 34989572 DOI: 10.1021/acs.chemrestox.1c00255] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mercury (Hg) is a toxic heavy-metal element, which can be enriched in fauna and flora and transformed into methylmercury (MeHg). MeHg is a widely distributed environmental pollutant that may be harmful to fish-eating populations through enrichment of aquatic food chains. The central nervous system is a primary target of MeHg. Embryos and infants are more sensitive to MeHg, and exposure to MeHg during gestational feeding can significantly impair the homeostasis of offspring, leading to long-term neurodevelopmental defects. At present, MeHg-induced neurodevelopmental toxicity has become a hotspot in the field of neurotoxicology, but its mechanisms are not fully understood. Some evidence point to oxidative damage, excitotoxicity, calcium ion imbalance, mitochondrial dysfunction, epigenetic changes, and other molecular mechanisms that play important roles in MeHg-induced neurodevelopmental toxicity. In this review, advances in the study of neurodevelopmental toxicity of MeHg exposure during pregnancy and the molecular mechanisms of related pathways are summarized, in order to provide more scientific basis for the study of neurodevelopmental toxicity of MeHg.
Collapse
Affiliation(s)
- Jingjing Pan
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Xiaoyang Li
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yanfeng Wei
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Linlin Ni
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Tianyao Yang
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, No.77 Puhe Road, Shenbei New District, Shenyang 110122, Liaoning China
| |
Collapse
|
31
|
Lamarche J, Alcoceba Álvarez E, Cordeau E, Enjalbal C, Massai L, Messori L, Lobinski R, Ronga L. Comparative reactivity of medicinal gold(I) compounds with the cyclic peptide vasopressin and its diselenide analogue. Dalton Trans 2021; 50:17487-17490. [PMID: 34796892 DOI: 10.1039/d1dt03470g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The reactions of the medicinal gold(I) compound auranofin and its close analogues with vasopressin and the diselenide analogue were comparatively investigated by LC-electrospray MS/MS. Evidence is gained of the possible cleavage of the S-S and Se-Se bridges induced by Au(I). Notably, we found that, in the absence of reducing agents, the sulfur and selenium atoms are metallated only at high temperature (70 °C) with the preferential binding of gold to selenium. The reaction with the S-S bridge can take place at physiological temperature (37 °C) under reducing conditions. The implications of these results are discussed in the general frame of the reactivity of biologically relevant soft Lewis acids with peptides and proteins.
Collapse
Affiliation(s)
- Jeremy Lamarche
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France.
| | | | | | | | - Lara Massai
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Luigi Messori
- Department of Chemistry, University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Ryszard Lobinski
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France. .,IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Luisa Ronga
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, Pau, France.
| |
Collapse
|
32
|
Tolbatov I, Marzo T, Coletti C, La Mendola D, Storchi L, Re N, Marrone A. Reactivity of antitumor coinage metal-based N-heterocyclic carbene complexes with cysteine and selenocysteine protein sites. J Inorg Biochem 2021; 223:111533. [PMID: 34273714 DOI: 10.1016/j.jinorgbio.2021.111533] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/17/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
The reaction of the antitumor M(I)-bis-N-heterocyclic carbene (M(I)-NHC) complexes, M = Cu, Ag, and Au, with their potential protein binding sites, i.e. cysteine and selenocysteine, was investigated by means of density functional theory approaches. Capped cysteine and selenocysteine were employed to better model the corresponding residues environment within peptide structures. By assuming the neutral or deprotonated form of the side chains of these amino acids and by considering the possible assistance of an external proton donor such as an adjacent acidic residue or the acidic component of the surrounding buffer environment, we devised five possible routes leading to the binding of the investigated M(I)-NHC scaffolds to these protein sites, reflecting their different location in the protein structure and exposure to the bulk. The targeting of either cysteine or selenocysteine in their neutral forms is a kinetically unfavored process, expected to be quite slow if observable at all at physiological temperature. On the other hand, the reaction with the deprotonated forms is much more favored, even though an external proton source is required to assist the protonation of the leaving carbene. Our calculations also show that all coinage metals are characterized by a similar reactivity toward the binding of cysteine and selenocysteine sites, although the Au(I) complex has significantly higher reaction and activation free energies compared to Cu(I) and Ag(I).
Collapse
Affiliation(s)
- Iogann Tolbatov
- Institut de Chimie Moleculaire de l'Université de Bourgogne (ICMUB), UMR CNRS 6302, Université de Bourgogne Franche-Comté(UBFC), Avenue Alain Savary 9, 21078 Dijon, France; Dipartimento di Farmacia, Università "G d'Annunzio" Chieti-Pescara, Via dei Vestini, Chieti, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; CISUP - Centre for Instrumentation Sharing (Centro per l'Integrazione della Strumentazione Scientifica), University of Pisa, Italy; University Consortium for Research in the Chemistry of Metal ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
| | - Cecilia Coletti
- Dipartimento di Farmacia, Università "G d'Annunzio" Chieti-Pescara, Via dei Vestini, Chieti, Italy.
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; University Consortium for Research in the Chemistry of Metal ions in Biological Systems (CIRCMSB), Via Celso Ulpiani 27, 70126 Bari, Italy
| | - Loriano Storchi
- Dipartimento di Farmacia, Università "G d'Annunzio" Chieti-Pescara, Via dei Vestini, Chieti, Italy
| | - Nazzareno Re
- Dipartimento di Farmacia, Università "G d'Annunzio" Chieti-Pescara, Via dei Vestini, Chieti, Italy
| | - Alessandro Marrone
- Dipartimento di Farmacia, Università "G d'Annunzio" Chieti-Pescara, Via dei Vestini, Chieti, Italy
| |
Collapse
|
33
|
Nehzati S, Dolgova NV, James AK, Cotelesage JJH, Sokaras D, Kroll T, George GN, Pickering IJ. High Energy Resolution Fluorescence Detected X-ray Absorption Spectroscopy: An Analytical Method for Selenium Speciation. Anal Chem 2021; 93:9235-9243. [PMID: 34164981 DOI: 10.1021/acs.analchem.1c01503] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Selenium is in many ways an enigmatic element. It is essential for health but toxic in excess, with the difference between the two doses being narrower than for any other element. Environmentally, selenium is of concern due to its toxicity. As the rarest of the essential elements, its low levels often provide challenges to the analytical chemist. X-ray absorption spectroscopy (XAS) provides a powerful tool for in situ chemical speciation but is severely limited by poor spectroscopic resolution arising from core-hole lifetime broadening. Here we explore selenium Kα1 high energy resolution fluorescence detected XAS (HERFD-XAS) as a novel approach for chemical speciation of selenium, in comparison with conventional Se K-edge XAS. We present spectra of a range of selenium species relevant to environmental and life science studies, including spectra of seleno-amino acids, which show strong similarities with S K-edge XAS of their sulfur congeners. We discuss strengths and limitations of HERFD-XAS, showing improvements in both speciation performance and low concentration detection. We also develop a simple method to correct fluorescence self-absorption artifacts, which is generally applicable to any HERFD-XAS experiment.
Collapse
Affiliation(s)
- Susan Nehzati
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Natalia V Dolgova
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Ashley K James
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada.,Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada
| | - Julien J H Cotelesage
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Graham N George
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada.,Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Ingrid J Pickering
- Molecular and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada.,Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5B3, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
34
|
Nogara PA, Madabeni A, Bortoli M, Teixeira Rocha JB, Orian L. Methylmercury Can Facilitate the Formation of Dehydroalanine in Selenoenzymes: Insight from DFT Molecular Modeling. Chem Res Toxicol 2021; 34:1655-1663. [PMID: 34077192 DOI: 10.1021/acs.chemrestox.1c00073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Experimental studies have indicated that electrophilic mercury forms (e.g., methylmercury, MeHg+) can accelerate the breakage of selenocysteine in vitro. Particularly, in 2009, Khan et al. (Environ. Toxicol. Chem. 2009, 28, 1567-1577) proposed a mechanism for the degradation of a free methylmercury selenocysteinate complex that was theoretically supported by Asaduzzaman et al. (Inorg. Chem. 2010, 50, 2366-2372). However, little is known about the fate of methylmercury selenocysteinate complexes embedded in an enzyme, especially in conditions of oxidative stress in which methylmercury target enzymes operate. Here, an accurate computational study on molecular models (level of theory: COSMO-ZORA-BLYP-D3(BJ)/TZ2P) was carried out to investigate the formation of dehydroalanine (Dha) in selenoenzymes, which irreversibly impairs their function. Methylselenocysteine as well as methylcysteine and methyltellurocysteine were included to gain insight on the peculiar behavior of selenium. Dha forms in a two-step process, i.e., the oxidation of the chalcogen nucleus followed by a syn-elimination leading to the alkene and the chalcogenic acid. The effect of an excess of hydrogen peroxide, which may lead to the formation of chalcogenones before the elimination, and of MeHg+, a severe toxicant targeting selenoproteins, which leads to the formation of methylmercury selenocysteinate, are also studied with the aim of assessing whether these pathological conditions facilitate the formation of Dha. Indeed, elimination occurs after chalcogen oxidation and MeHg+ facilitates the process. These results indicate a possible mechanism of toxicity of MeHg+ in selenoproteins.
Collapse
Affiliation(s)
- Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil.,Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Andrea Madabeni
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Marco Bortoli
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria 97105-900, RS, Brazil
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
35
|
Rajapaksha AA, Fu YX, Guo WY, Liu SY, Li ZW, Xiong CQ, Yang WC, Yang GF. Review on the recent progress in the development of fluorescent probes targeting enzymes. Methods Appl Fluoresc 2021; 9. [PMID: 33873170 DOI: 10.1088/2050-6120/abf988] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Enzymes are very important for biological processes in a living being, performing similar or multiple tasks in and out of cells, tissues and other organisms at a particular location. The abnormal activity of particular enzyme usually caused serious diseases such as Alzheimer's disease, Parkinson's disease, cancers, diabetes, cardiovascular diseases, arthritis etc. Hence, nondestructive and real-time visualization for certain enzyme is very important for understanding the biological issues, as well as the drug administration and drug metabolism. Fluorescent cellular probe-based enzyme detectionin vitroandin vivohas become broad interest for human disease diagnostics and therapeutics. This review highlights the recent findings and designs of highly sensitive and selective fluorescent cellular probes targeting enzymes for quantitative analysis and bioimaging.
Collapse
Affiliation(s)
- Asanka Amith Rajapaksha
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China.,Department of Nano Science Technology, Faculty of Technology, Wayamba University of Sri Lanka, Kuliyapitiya, Sri Lanka
| | - Yi-Xuan Fu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Wu Yingzheng Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Shi-Yu Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Zhi-Wen Li
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Cui-Qin Xiong
- Department of Interventional Medicine, Wuhan Third Hospital-Tongren Hospital of Wuhan University, Wuhan 430070, People's Republic of China
| | - Wen-Chao Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, People's Republic of China
| |
Collapse
|
36
|
Yaakoub H, Staerck C, Mina S, Godon C, Fleury M, Bouchara JP, Calenda A. Repurposing of auranofin and honokiol as antifungals against Scedosporium species and the related fungus Lomentospora prolificans. Virulence 2021; 12:1076-1090. [PMID: 33825667 PMCID: PMC8032236 DOI: 10.1080/21505594.2021.1909266] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The slowing-down de novo drug-discovery emphasized the importance of repurposing old drugs. This is particularly true when combating infections caused by therapy-refractory microorganisms, such as Scedosporium species and Lomentospora prolificans. Recent studies on Scedosporium responses to oxidative stress underscored the importance of targeting the underlying mechanisms. Auranofin, ebselen, PX-12, honokiol, and to a lesser extent, conoidin A are known to disturb redox-homeostasis systems in many organisms. Their antifungal activity was assessed against 27 isolates belonging to the major Scedosporium species: S. apiospermum, S. aurantiacum, S. boydii, S. dehoogii, S. minutisporum, and Lomentospora prolificans. Auranofin and honokiol were the most active against all Scedosporium species (mean MIC50 values of 2.875 and 6.143 μg/ml, respectively) and against L. prolificans isolates (mean MIC50 values of 4.0 and 3.563μg/ml respectively). Combinations of auranofin with voriconazole or honokiol revealed additive effects against 9/27 and 18/27 isolates, respectively. Synergistic interaction between auranofin and honokiol was only found against one isolate of L. prolificans. The effects of auranofin upon exposure to oxidative stress were also investigated. For all species except S. dehoogii, the maximal growth in the presence of auranofin significantly decreased when adding a sublethal dose of menadione. The analysis of the expression of genes encoding oxidoreductase enzymes upon exposure of S. apiospermum to honokiol unveiled the upregulation of many genes, especially those coding peroxiredoxins, thioredoxin reductases, and glutaredoxins. Altogether, these data suggest that auranofin and honokiol act via dampening the redox balance and support their repurposing as antifungals against Scedosporium species and L. prolificans.
Collapse
Affiliation(s)
- Hajar Yaakoub
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Cindy Staerck
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Sara Mina
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Charlotte Godon
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Maxime Fleury
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| | - Jean-Philippe Bouchara
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France.,Département de biologie des agents infectieux , Laboratoire De Parasitologie-Mycologie, Centre Hospitalier Universitaire, Angers, France
| | - Alphonse Calenda
- Groupe d'Etude Des Interactions Hôte-Pathogène (GEIHP, EA 3142), SFR ICAT 4208, UNIV Angers, UNIV Brest, Institut De Biologie En Santé-IRIS, CHU Angers,Angers, France
| |
Collapse
|
37
|
Madabeni A, Nogara PA, Bortoli M, Rocha JB, Orian L. Effect of Methylmercury Binding on the Peroxide-Reducing Potential of Cysteine and Selenocysteine. Inorg Chem 2021; 60:4646-4656. [PMID: 33587617 PMCID: PMC8763373 DOI: 10.1021/acs.inorgchem.0c03619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Indexed: 01/09/2023]
Abstract
Methylmercury (CH3Hg+) binding to catalytically fundamental cysteine and selenocysteine of peroxide-reducing enzymes has long been postulated as the origin of its toxicological activity. Only very recently, CH3Hg+ binding to the selenocysteine of thioredoxin reductase has been directly observed [Pickering, I. J. Inorg. Chem., 2020, 59, 2711-2718], but the precise influence of the toxicant on the peroxide-reducing potential of such a residue has never been investigated. In this work, we employ state-of-the-art density functional theory calculations to study the reactivity of molecular models of the free and toxified enzymes. Trends in activation energies are discussed with attention to the biological consequences and are rationalized within the chemically intuitive framework provided by the activation strain model. With respect to the free, protonated amino acids, CH3Hg+ binding promotes oxidation of the S or Se nucleus, suggesting that chalcogenoxide formation might occur in the toxified enzyme, even if the actual rate of peroxide reduction is almost certainly lowered as suggested by comparison with fully deprotonated amino acids models.
Collapse
Affiliation(s)
- Andrea Madabeni
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Pablo A. Nogara
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
- Departamento
de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa
Maria, RS, Brazil
| | - Marco Bortoli
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| | - João B.
T. Rocha
- Departamento
de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), 97105-900 Santa
Maria, RS, Brazil
| | - Laura Orian
- Dipartimento
di Scienze Chimiche, Università degli
Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
38
|
Manceau A, Bourdineaud JP, Oliveira RB, Sarrazin SLF, Krabbenhoft DP, Eagles-Smith CA, Ackerman JT, Stewart AR, Ward-Deitrich C, Del Castillo Busto ME, Goenaga-Infante H, Wack A, Retegan M, Detlefs B, Glatzel P, Bustamante P, Nagy KL, Poulin BA. Demethylation of Methylmercury in Bird, Fish, and Earthworm. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:1527-1534. [PMID: 33476127 DOI: 10.1021/acs.est.0c04948] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Toxicity of methylmercury (MeHg) to wildlife and humans results from its binding to cysteine residues of proteins, forming MeHg-cysteinate (MeHgCys) complexes that hinder biological functions. MeHgCys complexes can be detoxified in vivo, yet how this occurs is unknown. We report that MeHgCys complexes are transformed into selenocysteinate [Hg(Sec)4] complexes in multiple animals from two phyla (a waterbird, freshwater fish, and earthworms) sampled in different geographical areas and contaminated by different Hg sources. In addition, high energy-resolution X-ray absorption spectroscopy (HR-XANES) and chromatography-inductively coupled plasma mass spectrometry of the waterbird liver support the binding of Hg(Sec)4 to selenoprotein P and biomineralization of Hg(Sec)4 to chemically inert nanoparticulate mercury selenide (HgSe). The results provide a foundation for understanding mercury detoxification in higher organisms and suggest that the identified MeHgCys to Hg(Sec)4 demethylation pathway is common in nature.
Collapse
Affiliation(s)
- Alain Manceau
- Université Grenoble Alpes, ISTerre, CNRS, Grenoble 38000, France
| | - Jean-Paul Bourdineaud
- Université de Bordeaux, Institut Européen de Chimie et Biologie, CNRS, Pessac 33600, France
| | - Ricardo B Oliveira
- Universidade Federal do Oeste Pará, LabBBEx, Santarém 68180-000, Pará, Brazil
| | - Sandra L F Sarrazin
- Universidade Federal do Oeste Pará, LabBBEx, Santarém 68180-000, Pará, Brazil
| | - David P Krabbenhoft
- Upper Midwest Water Science Center, U.S. Geological Survey, Middleton 53562, Wisconsin, United States
| | - Collin A Eagles-Smith
- Forest and Rangeland Ecosystem Science Center, U.S. Geological Survey, Corvallis 97330, Oregon, United States
| | - Joshua T Ackerman
- Western Ecological Research Center, U.S. Geological Survey, Dixon Field Station, Dixon 95620, California, United States
| | - A Robin Stewart
- U.S. Geological Survey, Water Resources Mission Area, Menlo Park 94025, California, United States
| | | | | | | | - Aude Wack
- Université Grenoble Alpes, ISTerre, CNRS, Grenoble 38000, France
| | - Marius Retegan
- European Synchrotron Radiation Facility (ESRF), Grenoble 38000, France
| | - Blanka Detlefs
- European Synchrotron Radiation Facility (ESRF), Grenoble 38000, France
| | - Pieter Glatzel
- European Synchrotron Radiation Facility (ESRF), Grenoble 38000, France
| | - Paco Bustamante
- Université La Rochelle, CNRS, Littoral Environnement et Sociétés, La Rochelle 17000, France
| | - Kathryn L Nagy
- Department of Earth and Environmental Sciences, University of Illinois at Chicago, Chicago 60607, Illinois, United States
| | - Brett A Poulin
- U.S. Geological Survey, Water Resources Mission Area, Boulder 80303, Colorado, United States
- Department of Environmental Toxicology, University of California Davis, Davis 95616, California, United States
| |
Collapse
|
39
|
Zoppi C, Messori L, Pratesi A. ESI MS studies highlight the selective interaction of Auranofin with protein free thiols. Dalton Trans 2020; 49:5906-5913. [PMID: 32314767 DOI: 10.1039/d0dt00283f] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The clinically established gold drug Auranofin was reacted individually with a group of representative proteins, namely ubiquitin, ribonuclease A, carbonic anhydrase, haemoglobin and superoxide dismutase, and adduct formation was monitored in the various cases by ESI-MS analysis. We found that the reaction is highly selective for solvent exposed free cysteines that are modified through coordination of the AuPEt3+ fragment. Indeed, ESI-Q-TOF MS spectra carried out on protein samples incubated with a three fold molar excess of Auranofin allowed direct detection of the native proteins bearing bound AuPEt3+ fragments in the cases of carbonic anhydrase and haemoglobin. At variance, the two proteins that do not possess any free cysteine residue, i.e. ubiquitin and ribonuclease A, were unable to bind the gold fragment. In the case of superoxide dismutase, adduct formation is hindered by the scarce solvent accessibility of the free cysteine residue. These findings were further confirmed by a series of competition binding experiments with ebselen, a potent and selective cysteine-modifying reagent; we observed that pre-treatment with ebselen prevents the binding of the AuPEt3+ fragment to both carbonic anhydrase and haemoglobin.
Collapse
Affiliation(s)
- Carlotta Zoppi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, Italy.
| | | | | |
Collapse
|
40
|
Clapper E, Wang S, Raninga PV, Di Trapani G, Tonissen KF. Cross-talk between Bcr-abl and the Thioredoxin System in Chronic Myeloid Leukaemia: Implications for CML Treatment. Antioxidants (Basel) 2020; 9:E207. [PMID: 32138149 PMCID: PMC7139888 DOI: 10.3390/antiox9030207] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic myeloid leukaemia (CML) is currently treated with inhibitors of the CML specific oncoprotein, bcr-abl. While this strategy is initially successful, drug resistance can become a problem. Therefore, new targets need to be identified to ensure the disease can be appropriately managed. The thioredoxin (Trx) system, comprised of Trx, thioredoxin reductase (TrxR), and NADPH, is an antioxidant system previously identified as a target for therapies aimed at overcoming drug resistance in other cancers. We assessed the effectiveness of TrxR inhibitors on drug resistant CML cells and examined links between TrxR and the bcr-abl cell-signalling pathway. Two TrxR inhibitors, auranofin and [Au(d2pype)2]Cl, increased intracellular ROS levels and elicited apoptosis in both sensitive and imatinib resistant CML cells. Inhibition of TrxR activity by these pharmacological inhibitors, or by specific siRNA, also resulted in decreased bcr-abl mRNA and protein levels, and lower bcr-abl downstream signalling activity, potentially enhancing the effectiveness of TrxR inhibitors as CML therapies. In addition, imatinib resistant CML cell lines showed upregulated expression of the Trx system. Furthermore, analysis of datasets showed that CML patients who did not respond to imatinib had higher Trx mRNA levels than patients who responded to treatment. Our study demonstrates a link between the Trx system and the bcr-abl protein and highlights the therapeutic potential of targeting the Trx system to improve CML patients' outcomes.
Collapse
Affiliation(s)
- Erin Clapper
- School of Environment and Science, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (E.C.); (S.W.)
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia
| | - Sicong Wang
- School of Environment and Science, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (E.C.); (S.W.)
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia
| | - Prahlad V. Raninga
- Signal Transduction Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia;
| | - Giovanna Di Trapani
- School of Environment and Science, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (E.C.); (S.W.)
| | - Kathryn F. Tonissen
- School of Environment and Science, Griffith University, Nathan, Brisbane, QLD 4111, Australia; (E.C.); (S.W.)
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, QLD 4111, Australia
| |
Collapse
|