1
|
Rehem AR, da Gama Viveiro LR, De Souza Santos EL, do Carmo PHF, da Silva NS, Junqueira JC, Scorzoni L. Antifungal and antibiofilm effect of duloxetine hydrochloride against Cryptococcus neoformans and Cryptococcus gattii. Folia Microbiol (Praha) 2024; 69:1247-1254. [PMID: 38652436 DOI: 10.1007/s12223-024-01164-1] [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: 10/24/2023] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Cryptococcosis is an invasive mycosis caused mainly by Cryptococcus gattii and C. neoformans and is treated with amphotericin B (AMB), fluconazole and 5-fluorocytosine. However, antifungal resistance, limited and toxic antifungal arsenal stimulate the search for therapeutic strategies such as drug repurposing. Among the repurposed drugs studied, the selective serotonin reuptake inhibitors (SSRIs) have shown activity against Cryptococcus spp. However, little is known about the antifungal effect of duloxetine hydrochloride (DH), a selective serotonin and norepinephrine reuptake inhibitor (SSNRI), against C. neoformans and C. gattii. In this study, DH inhibited the growth of several C. neoformans and C. gattii strains at concentrations ranging from 15.62 to 62.50 µg/mL. In addition, DH exhibited fungicidal activity ranging from 15.62 to 250 µg/mL. In biofilm, DH treatment reduced Cryptococcus spp. biomass at a level comparable to AMB, with a significant reduction (85%) for C. neoformans biofilms. The metabolic activity of C. neoformans and C. gattii biofilms decreased significantly (99%) after treatment with DH. Scanning electron micrographs confirmed the anti-biofilm activity of DH, as isolated cells could be observed after treatment. In conclusion, DH showed promising antifungal activity against planktonic cells and biofilms of C. neoformans and C. gattii, opening perspectives for further studies with DH in vivo.
Collapse
Affiliation(s)
- Amanda Rodrigues Rehem
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Letícia Rampazzo da Gama Viveiro
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Evelyn Luzia De Souza Santos
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Paulo Henrique Fonseca do Carmo
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Newton Soares da Silva
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil
| | - Liliana Scorzoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), Av. Engenheiro Francisco José Longo, 777 São José dos Campos, São Paulo 12245-000, Brazil.
- Universidade de Guarulhos (UNG), Programa de Pós-Graduação em Enfermagem, Guarulhos, SP, Brasil.
| |
Collapse
|
2
|
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
|
3
|
Büssing R, Bublitz A, Karge B, Brönstrup M, Strowig T, Ott I. An organometallic hybrid antibiotic of metronidazole with a Gold(I) N-Heterocyclic Carbene overcomes metronidazole resistance in Clostridioides difficile. J Biol Inorg Chem 2024; 29:511-518. [PMID: 38926159 PMCID: PMC11343806 DOI: 10.1007/s00775-024-02064-y] [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: 03/01/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Antimicrobial resistance (AMR) has been emerging as a major global health threat and calls for the development of novel drug candidates. Metal complexes have been demonstrating high efficiency as antibacterial agents that differ substantially from the established types of antibiotics in their chemical structures and their mechanism of action. One strategy to exploit this potential is the design of metal-based hybrid organometallics that consist of an established antibiotic and a metal-based warhead that contributes an additional mechanism of action different from that of the parent antibiotic. In this communication, we describe the organometallic hybrid antibiotic 2c, in which the drug metronidazole is connected to a gold(I) N-heterocyclic carbene warhead that inhibits bacterial thioredoxin reductase (TrxR). Metronidazole can be used for the treatment with the obligatory anaerobic pathogen Clostridioides difficile (C. difficile), however, resistance to the drug hampers its clinical success. The gold organometallic conjugate 2c was an efficient inhibitor of TrxR and it was inactive or showed only minor effects against eucaryotic cells and bacteria grown under aerobic conditions. In contrast, a strong antibacterial effect was observed against both metronidazole-sensitive and -resistant strains of C. difficile. This report presents a proof-of-concept that the design of metal-based hybrid antibiotics can be a viable approach to efficiently tackle AMR.
Collapse
Affiliation(s)
- Rolf Büssing
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, 38106, Braunschweig, Germany
| | - Arne Bublitz
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Bianka Karge
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstr. 55, 38106, Braunschweig, Germany.
| |
Collapse
|
4
|
Mahdavi SM, Bockfeld D, Esarev IV, Lippmann P, Frank R, Brönstrup M, Ott I, Tamm M. Gold(i) and gold(iii) carbene complexes from the marine betaine norzooanemonin: inhibition of thioredoxin reductase, antiproliferative and antimicrobial activity. RSC Med Chem 2024:d4md00358f. [PMID: 39185451 PMCID: PMC11342128 DOI: 10.1039/d4md00358f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
The natural marine betaine norzooanemonin (1,3-dimethylimidazolim-4-carboxylate) and its methyl and ethyl esters were used as ligand precursors to prepare a systematic series (12 members) of neutral monocarbene gold(i/iii) and cationic dicarbene gold(i/iii) complexes. The complexes were evaluated as inhibitors of bacterial thioredoxin reductase and for their antiproliferative and antimicrobial activities. While gold complexes with the parent norzooanemonin scaffold resulted in overall poor performance, the more lipophilic esters proved to be highly bioactive agents, related to their higher cellular uptake. The monocarbene gold(i/iii) complexes showed significant potency as inhibitors of bacterial thioredoxin reductase. In most assays, the efficacy of both gold(i) and gold(iii) analogues was found to be comparable. The cytotoxicity of dicarbene gold(i/iii) complexes against cancer cells was strong, in some cases exceeding that of the standard reference auranofin.
Collapse
Affiliation(s)
- Seyedeh Mahbobeh Mahdavi
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig Hagenring30 38106 Braunschweig Germany
| | - Dirk Bockfeld
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig Hagenring30 38106 Braunschweig Germany
| | - Igor V Esarev
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstraße 55 38106 Braunschweig Germany
| | - Petra Lippmann
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstraße 55 38106 Braunschweig Germany
| | - René Frank
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig Hagenring30 38106 Braunschweig Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research GmbH Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstraße 55 38106 Braunschweig Germany
| | - Matthias Tamm
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig Hagenring30 38106 Braunschweig Germany
| |
Collapse
|
5
|
Coscione F, Zineddu S, Vitali V, Fondi M, Messori L, Perrin E. The Many Lives of Auranofin: How an Old Anti-Rheumatic Agent May Become a Promising Antimicrobial Drug. Antibiotics (Basel) 2024; 13:652. [PMID: 39061334 PMCID: PMC11274207 DOI: 10.3390/antibiotics13070652] [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: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases.
Collapse
Affiliation(s)
- Francesca Coscione
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Stefano Zineddu
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Valentina Vitali
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Marco Fondi
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy; (S.Z.); (V.V.)
| | - Elena Perrin
- Department of Biology, University of Florence, Via Madonna del Piano 6, I-50019 Sesto Fiorentino, Italy; (F.C.); (M.F.)
| |
Collapse
|
6
|
Vitali V, Massai L, Messori L. Strategies for the design of analogs of auranofin endowed with anticancer potential. Expert Opin Drug Discov 2024; 19:855-867. [PMID: 38803122 DOI: 10.1080/17460441.2024.2355329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Auranofin (AF) is a well-established, FDA-approved, antiarthritic gold drug that is currently being reevaluated for a variety of therapeutic indications through drug repurposing. AF has shown great promise as a potential anticancer agent and has been approved for a few clinical trials in cancer. The renewed interest in AF has led to extensive research into the design, preparation and biological evaluation of auranofin analogs, which may have an even better pharmacological profile than the parent drug. AREAS COVERED This article reviews the strategies for chemical modification of the AF scaffold. Several auranofin analogs have been prepared and characterized for medical application in the field of cancer treatment over the last 20 years. Some emerging structure-function relationships are proposed and discussed. EXPERT OPINION The chemical modification of the AF scaffold has been the subject of intense activity in recent years and this strategy has led to the preparation and evaluation of several AF analogs. The case of iodauranofin is a particularly promising example. The availability of homogeneous biological data for a group of AF derivatives allows some initial structure-function relationships to be proposed, which may inspire the design and synthesis of new and better AF analogs for cancer treatment.
Collapse
Affiliation(s)
- Valentina Vitali
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
7
|
Marques A, Carabineiro SAC, Aureliano M, Faleiro L. Evaluation of Gold Complexes to Address Bacterial Resistance, Quorum Sensing, Biofilm Formation, and Their Antiviral Properties against Bacteriophages. TOXICS 2023; 11:879. [PMID: 37999531 PMCID: PMC10674251 DOI: 10.3390/toxics11110879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023]
Abstract
The worldwide increase in antibiotic resistance poses a significant challenge, and researchers are diligently seeking new drugs to combat infections and prevent bacterial pathogens from developing resistance. Gold (I and III) complexes are suitable for this purpose. In this study, we tested four gold (I and III) complexes, (1) chlorotrimethylphosphine gold(I); (2) chlorotriphenylphosphine gold(I); (3) dichloro(2-pyridinecarboxylate) gold (III); and (4) 1,3-bis(2,6-diisopropylphenyl)imidazole-2-ylidene gold(I) chloride, for their antibacterial, antibiofilm, antiviral, and anti-quorum sensing activities. Results reveal that 1 significantly inhibits Escherichia coli DSM 1077 and Staphylococcus aureus ATCC 6538, while 2, 3, and 4 only inhibit S. aureus ATCC 6538. The minimum inhibitory concentration (MIC) of 1 for S. aureus ATCC 6538 is 0.59 μg/mL (1.91 μM), and for methicillin-resistant S. aureus strains MRSA 12 and MRSA 15, it is 1.16 μg/mL (3.75 μM). For E. coli DSM 1077 (Gram-negative), the MIC is 4.63 μg/mL (15 μM), and for multi-resistant E. coli I731940778-1, it is 9.25 μg/mL (30 μM). Complex 1 also disrupts biofilm formation in E. coli and S. aureus after 6 h or 24 h exposure. Moreover, 1 and 2 inhibit the replication of two enterobacteria phages. Anti-quorum sensing potential still requires further clarification. These findings highlight the potential of gold complexes as effective agents to combat bacterial and viral infections.
Collapse
Affiliation(s)
- Ana Marques
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
- Algarve Biomedical Center—Research Institute, 8005-139 Faro, Portugal
| | - Sónia A. C. Carabineiro
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal;
| | - Manuel Aureliano
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
- Centro de Ciências do Mar (CCMar), Universidade do Algarve, 8005-139 Faro, Portugal
| | - Leonor Faleiro
- Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal;
- Algarve Biomedical Center—Research Institute, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
8
|
de Andrade Querino AL, de Sousa AM, Thomas SR, de Lima GM, Dittz D, Casini A, do Monte-Neto RL, Silva H. Organogold(III)-dithiocarbamate compounds and their coordination analogues as anti-tumor and anti-leishmanial metallodrugs. J Inorg Biochem 2023; 247:112346. [PMID: 37536162 DOI: 10.1016/j.jinorgbio.2023.112346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
The limited chemical stability of gold(III)-based compounds in physiological environment has been a challenge in drug discovery, and organometallic chemistry might provide the solution to overcome this issue. In this work, four novel cationic organogold(III)-dithiocarbamate complexes of general structure [(C^N)AuIIIDTC]PF6 (C1a - C4a, DTC = dithiocarbamate, L1 - L4, C^N = 2-anilinopyridine) are presented, and compared to their coordination gold(III)-dithiocarbamate analogues [AuIIIDTCCl2] (C1b - C4b), as potential anti-cancer and anti-leishmanial drugs. Most of the complexes effectively inhibited cancer cell growth, notably C3a presented anti-proliferative effect in the nanomolar range against breast cancer (MCF-7 and MDA-MB-231 cells with moderate selectivity. Pro-apoptotic studies on treated MCF-7 cells showed a high population of cells in early apoptosis. Reactivity studies of C3a towards model thiols (N-acetyl-L-cysteine) refer to a possible mode of action involving bonding between the organogold(III)-core and the thiolate. In the scope of neglected diseases, gold complexes are emerging as promising therapeutic alternatives against leishmaniasis. In this regard, all gold(III)-dithiocarbamate complexes presented anti-leishmanial activity against at least one Leishmania species. Complexes C1a, C4a, C1b, C4b were active against all tested parasites with IC50 values varying between 0.12 and 42 μM, and, overall, organometallic compounds presented more intriguing inhibition profiles. For C4a selectivity over 500-fold for L. braziliensis; even higher than the reference anti-leishmanial drug amphotericin B. Overall, our findings revealed that the organogold(III) moiety significantly amplified the anti-cancer and anti-leishmanial effects with respect to the coordination analogues; thus, showing the great potential of organometallic chemistry in metallodrug-based chemotherapy for cancer and leishmaniasis.
Collapse
Affiliation(s)
- Ana Luiza de Andrade Querino
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany.
| | - Alessandra Mara de Sousa
- RdM Lab - Biotechnology Applied to Pathogens Research Group, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Sophie R Thomas
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany
| | - Geraldo Magela de Lima
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dalton Dittz
- Department of Biochemistry and Pharmacology, Universidade Federal do Piaui, Teresina, Brazil
| | - Angela Casini
- Chair of Medicinal and Bioinorganic Chemistry, Department of Chemistry, Technical University of Munich, Garching b. Munich, Germany
| | - Rubens Lima do Monte-Neto
- RdM Lab - Biotechnology Applied to Pathogens Research Group, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Heveline Silva
- Laboratório de Síntese e Interações Bioinorgânicas (SibLab), Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Kaur M, Thakare R, Bhattacherya A, Murugan PA, Kaul G, Shukla M, Singh AK, Matheshwaran S, Chopra S, Bera JK. Antimicrobial efficacy of a hemilabile Pt(II)-NHC compound against drug-resistant S. aureus and Enterococcus. Dalton Trans 2023; 52:1876-1884. [PMID: 36648294 DOI: 10.1039/d2dt03365h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Three platinum(II)-N-heterocyclic carbene (NHC) compounds [Pt(L1)Cl](PF6) (1), [Pt(L2)(COD)](PF6)2 (2) and [Pt(L2)Cl2] (3) were synthesized bearing pyridyl-functionalized butenyl-tethered (L1H) and n-butyl tethered (L2H) NHC ligands, and their antibacterial activity against clinically relevant human pathogens was evaluated. Complex 1 was designed to have one of its metal coordination sites masked with a hemilabile butenyl group. The antibacterial activity spectrum against the ESKAPE panel of pathogens shows superior activity of 1 compared to 2 and 3 against the Gram-positive S. aureus pathogen. Complex 1 showed equipotent activity against clinical drug-resistant S. aureus and Enterococcus isolates. Furthermore, 1 demonstrated concentration-dependent bactericidal activity with a long post-antibiotic effect, eradicated preformed S. aureus biofilm and synergized with gentamicin and minocycline for combinatorial antimicrobial therapy. Under in vivo conditions, 1 displayed potent activity in reducing bacterial load in a murine thigh infection model, similar to vancomycin, albeit at 2.5× less dosage. An array of experiments reveals key characteristics for the hemilabile complex 1 as a potential anti-staphylococcal drug.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Chemistry and Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Ritesh Thakare
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow-226031, India.
| | - Arindom Bhattacherya
- Department of Chemistry and Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Prem Anand Murugan
- Department of Biological Sciences and Bioengineering, Centre for Environmental Sciences and Engineering, and Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Grace Kaul
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow-226031, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manjulika Shukla
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow-226031, India.
| | - Alok Kr Singh
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow-226031, India. .,Current Affiliation: Amity Institute of Biotechnology, Amity University Noida Campus, Sector-125, Noida - 201313, U.P., India
| | - Saravanan Matheshwaran
- Department of Biological Sciences and Bioengineering, Centre for Environmental Sciences and Engineering, and Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Sidharth Chopra
- Department of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow-226031, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jitendra K Bera
- Department of Chemistry and Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| |
Collapse
|
12
|
Antitumoral and Antimicrobial Activities of Block Copolymer Micelles Containing Gold Bisdithiolate Complexes. Pharmaceutics 2023; 15:pharmaceutics15020564. [PMID: 36839886 PMCID: PMC9964654 DOI: 10.3390/pharmaceutics15020564] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Gold(III) bisdithiolate complexes have been reported as potential antimicrobial and antitumoral agents. The complex [Au(cdc)2]- (cdc=cyanodithioimido carbonate) displayed antimicrobial and outstanding antitumor activity against the ovarian cancer cells A2780 and A2780cisR, which are sensitive and resistant to cisplatin, respectively. However, poor water solubility may hamper its clinical use. Block copolymer micelles (BCMs) may solubilize hydrophobic drugs, improving their bioavailability and circulation time in blood. Aiming to provide water solubility, prolonged availability, and enhanced therapeutic indexes, BCMs loaded with [Au(cdc)2]- were synthesized and characterized. The BCM-[Au(cdc)2] micelles were prepared with a loading efficiency of 64.6% and a loading content of 35.3 mg [Au(cdc)2]-/gBCM. A hydrodynamic diameter of 77.31 ± 27.00 nm and a low polydispersity index of 0.18 indicated that the micelles were homogenous and good candidates for drug delivery. Cytotoxic activity studies against A2780/A2780cisR cells showed that BCM-[Au(cdc)2] maintained relevant cytotoxic activity comparable to the cytotoxicity observed for the same concentration of gold complexes. The Au uptake in A2780 cells, determined by PIXE, was ca. 17% higher for BCMs-[Au(cdc)2] compared to [Au(cdc)2]-. The BCMs-[Au(cdc)2] presented antimicrobial activity against S. aureus Newman and C. glabrata CBS138. These results evidenced the potential of BCM-[Au(cdc)2] for drug delivery and its promising anticancer and antimicrobial activities.
Collapse
|
13
|
Mármol I, Quero J, Azcárate P, Atrián-Blasco E, Ramos C, Santos J, Gimeno MC, Rodríguez-Yoldi MJ, Cerrada E. Biological Activity of NHC-Gold-Alkynyl Complexes Derived from 3-Hydroxyflavones. Pharmaceutics 2022; 14:pharmaceutics14102064. [PMID: 36297498 PMCID: PMC9612383 DOI: 10.3390/pharmaceutics14102064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
In this paper we describe the synthesis of new N-heterocyclic carbene (NHC) gold(I) derivatives with flavone-derived ligands with a propargyl ether group. The compounds were screened for their antimicrobial and anticancer activities, showing greater activity against bacteria than against colon cancer cells (Caco-2). Complexes [Au(L2b)(IMe)] (1b) and [Au(L2b)(IPr)] (2b) were found to be active against both Gram-positive and Gram-negative strains. The mechanism of action of 1b was evaluated by measurement of thioredoxin reductase (TrxR) and dihydrofolate reductase (DHFR) activity, besides scanning electron microscopy (SEM). Inhibition of the enzyme thioredoxin reductase is not observed in either Escherichia Coli or Caco-2 cells; however, DHFR activity is compromised after incubation of E. coli cells with complex 1b. Moreover, loss of structural integrity and change in bacterial shape is observed in the images obtained from scanning electron microscopy (SEM) after treatment E. coli cells with complex 1b.
Collapse
Affiliation(s)
- Inés Mármol
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Departamento de Farmacología y Fisiología, Medicina Legal y Forense, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013 Zaragoza, Spain
| | - Javier Quero
- Departamento de Farmacología y Fisiología, Medicina Legal y Forense, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013 Zaragoza, Spain
| | - Paula Azcárate
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Elena Atrián-Blasco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain
| | - Carla Ramos
- Escola Superior de Tecnologia e Gestão, Instituto Politécnico de Viana do Castelo, Avenida do Atlântico No. 644, 4900-348 Viana do Castelo, Portugal
| | - Joana Santos
- Escola Superior de Tecnologia e Gestão, Instituto Politécnico de Viana do Castelo, Avenida do Atlântico No. 644, 4900-348 Viana do Castelo, Portugal
| | - María Concepción Gimeno
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - María Jesús Rodríguez-Yoldi
- Departamento de Farmacología y Fisiología, Medicina Legal y Forense, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013 Zaragoza, Spain
- Correspondence: (M.J.R.-Y.); (E.C.)
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Correspondence: (M.J.R.-Y.); (E.C.)
| |
Collapse
|
14
|
Non-Antibiotic Drug Repositioning as an Alternative Antimicrobial Approach. Antibiotics (Basel) 2022; 11:antibiotics11060816. [PMID: 35740222 PMCID: PMC9220406 DOI: 10.3390/antibiotics11060816] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
The worldwide scenario of antibiotic resistance and the falling number of funds for the development of novel antibiotics have led research efforts toward the study of specific cost-effective strategies aimed at discovering drugs against microbial infections. Among the potential options, drug repositioning, which has already exhibited satisfactory results in other medical fields, came out as the most promising. It consists of finding new uses for previously approved medicines and, over the years, many “repurposed drugs” displayed some encouraging in vitro and in vivo results beyond their initial application. The principal theoretical justification for reusing already existing drugs is that they have known mechanisms of action and manageable side effects. Reuse of old drugs is now considered an interesting approach to overcome the drawbacks of conventional antibiotics. The purpose of this review is to offer the reader a panoramic view of the updated studies concerning the repositioning process of different classes of non-antibiotic drugs in the antimicrobial field. Several research works reported the ability of some non-steroidal anti-inflammatory drugs (NSAIDs), antidepressants, antipsychotics, and statins to counteract the growth of harmful microorganisms, demonstrating an interesting winning mode to fight infectious diseases caused by antimicrobial resistant bacteria.
Collapse
|
15
|
Chiappetta G, Gamberi T, Faienza F, Limaj X, Rizza S, Messori L, Filomeni G, Modesti A, Vinh J. Redox proteome analysis of auranofin exposed ovarian cancer cells (A2780). Redox Biol 2022; 52:102294. [PMID: 35358852 PMCID: PMC8966199 DOI: 10.1016/j.redox.2022.102294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023] Open
Abstract
The effects of Auranofin (AF) on protein expression and protein oxidation in A2780 cancer cells were investigated through a strategy based on simultaneous expression proteomics and redox proteomics determinations. Bioinformatics analysis of the proteomics data supports the view that the most critical cellular changes elicited by AF treatment consist of thioredoxin reductase inhibition, alteration of the cell redox state, impairment of the mitochondrial functions, metabolic changes associated with conversion to a glycolytic phenotype, induction of ER stress. The occurrence of the above cellular changes was extensively validated by performing direct biochemical assays. Our data are consistent with the concept that AF produces its effects through a multitarget mechanism that mainly affects the redox metabolism and the mitochondrial functions and results into severe ER stress. Results are discussed in the context of the current mechanistic knowledge existing on AF. Redox proteomics allows to underline cell adaptation mechanisms in response to Auranofin treatment in ovarian cancer cells. BRCA1 is one of the major candidates of the ovarian cancer cell adaptation to Auranofin treatment. Auranofin alters the oxidative phosphorylation and mitochondrial protein import machinery. TRAP1 C501 modulates Auranofin toxicity. Auranofin induces severe stress of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Fiorella Faienza
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Xhesika Limaj
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Luigi Messori
- Metmed Lab, Department of Chemistry, University of Florence, via della lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark; Center for Healthy Aging, University of Copenhagen, Denmark
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Joelle Vinh
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| |
Collapse
|
16
|
Liu Y, Lu Y, Xu Z, Ma X, Chen X, Liu W. Repurposing of the gold drug auranofin and a review of its derivatives as antibacterial therapeutics. Drug Discov Today 2022; 27:1961-1973. [DOI: 10.1016/j.drudis.2022.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/22/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022]
|
17
|
Büssing R, Karge B, Lippmann P, Jones PG, Brönstrup M, Ott I. Gold(I) and Gold(III) N-Heterocyclic Carbene Complexes as Antibacterial Agents and Inhibitors of Bacterial Thioredoxin Reductase. ChemMedChem 2021; 16:3402-3409. [PMID: 34268875 PMCID: PMC9290826 DOI: 10.1002/cmdc.202100381] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/10/2021] [Indexed: 12/14/2022]
Abstract
A series of (NHC)Au(I)Cl monocarbene complexes and their gold(III) analogues (NHC)Au(III)Cl3 were prepared and investigated as antibacterial agents and inhibitors of bacterial TrxR. The complexes showed stronger antibacterial effects against the Gram-positive MRSA and E. faecium strains than against several Gram-negative bacteria. All complexes were efficient inhibitors of bacterial thioredoxin reductase, indicating that inhibition of this enzyme might be involved in their mechanism of action. The efficacy of gold(I) and gold(III) analogues was comparable in most of the assays. The cytotoxicity of the gold NHC compounds against cancer and human cells was overall weaker than the activity against the Gram-positive bacteria, suggesting that their optimization as antibacterials warrants further investigation.
Collapse
Affiliation(s)
- Rolf Büssing
- Institute of Medicinal and Pharmaceutical ChemistryTechnische Universität BraunschweigBeethovenstrasse 5538106BraunschweigGermany
| | - Bianka Karge
- Department of Chemical BiologyHelmholtz Centre for Infection Research and German Centre for Infection Research (DZIF)Inhoffenstrasse 738124BraunschweigGermany
| | - Petra Lippmann
- Institute of Medicinal and Pharmaceutical ChemistryTechnische Universität BraunschweigBeethovenstrasse 5538106BraunschweigGermany
| | - Peter G. Jones
- Institute of Inorganic and Analytical ChemistryTechnische Universität BraunschweigHagenring 3038106BraunschweigGermany
| | - Mark Brönstrup
- Department of Chemical BiologyHelmholtz Centre for Infection Research and German Centre for Infection Research (DZIF)Inhoffenstrasse 738124BraunschweigGermany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical ChemistryTechnische Universität BraunschweigBeethovenstrasse 5538106BraunschweigGermany
| |
Collapse
|
18
|
Mazzei L, Massai L, Cianci M, Messori L, Ciurli S. Medicinal Au(I) compounds targeting urease as prospective antimicrobial agents: unveiling the structural basis for enzyme inhibition. Dalton Trans 2021; 50:14444-14452. [PMID: 34585201 DOI: 10.1039/d1dt02488d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A few gold compounds were recently found to show antimicrobial properties in vitro, holding great promise for the discovery of new drugs to overcome antibiotic resistance. Here, the inhibition of the bacterial virulence factor urease by four Au(I)-compounds, namely Au(PEt3)Cl, Au(PEt3)Br, Au(PEt3)I and [Au(PEt3)2]Cl, obtained from the antiarthritic Au(I)-drug Auranofin and earlier reported to act as antimicrobials, is investigated. The three monophosphino Au(I) complexes showed IC50 values in the 30-100 nM range, while the diphosphino Au(I) complex, though being less active, still showed a IC50 value of 7 μM. The structural basis for this inhibition was provided by solving the crystal structures of urease co-crystallized with Au(PEt3)I and [Au(PEt3)2]Cl: at least two Au(I) ions bind the enzyme in a flap domain involved in the catalysis, thus obliterating enzyme activity. Peculiar changes observed in the two structures reveal implications for the mechanism of soft metal binding and enzyme inactivation.
Collapse
Affiliation(s)
- Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Giuseppe Fanin 40, I-40127 Bologna, Italy.
| | - Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, I-60131 Ancona, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, I-50019 Sesto Fiorentino, Italy
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Giuseppe Fanin 40, I-40127 Bologna, Italy.
| |
Collapse
|
19
|
Matiadis D, Stefanou V, Tsironis D, Panagiotopoulou A, Igglessi-Markopoulou O, Markopoulos J. Synthesis and preliminary biological evaluation of antibacterial and antifungal 5-arylidene tetramic acid-cadmium(II) complexes. Arch Pharm (Weinheim) 2021; 354:e2100305. [PMID: 34570387 DOI: 10.1002/ardp.202100305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 11/09/2022]
Abstract
The synthesis and biological evaluation of 5-arylidene-N-acetyl-tetramic acids cadmium(II) complexes are reported. Eleven novel compounds were prepared, characterized by nuclear magnetic resonance experiments and screened for their antimicrobial activity against five bacterial species (Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus [MRSA]) and two fungi (Candida albicans and Cryptococcus neoformans). The complexes showed similar or enhanced activities against MRSA in comparison to the corresponding ligands and, additionally, promising antifungal activities against C. neoformans. The most active compounds 3c and 3h showed remarkable activities against MRSA (minimum inhibitory activity [MIC] values of 32 and 4 μg/ml, respectively) and C. neoformans (MIC values of 8 and 16 μg/ml, respectively), accompanied by no human cell toxicity and hemolytic activity within the tested concentration range. The results demonstrate that appropriately functionalized tetramic acids attached with lipophilic alkanoyl chain and after complexation with cadmium(II) ions may act as valuable lead compounds for further investigations toward the development of novel antibacterial and/or antifungal agents.
Collapse
Affiliation(s)
- Dimitris Matiadis
- Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, Athens, Greece
| | - Valentina Stefanou
- Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, Athens, Greece
| | - Dimitrios Tsironis
- Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, Athens, Greece
| | - Angeliki Panagiotopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Ag. Paraskevi, Attiki, Greece
| | - Olga Igglessi-Markopoulou
- Laboratory of Organic Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, Athens, Greece
| | - John Markopoulos
- Department of Chemistry, Laboratory of Inorganic Chemistry, University of Athens, Panepistimiopolis, Athens, Greece
| |
Collapse
|
20
|
Hutton ML, Pehlivanoglu H, Vidor CJ, James ML, Thomson MJ, Lyras D. Repurposing auranofin as a Clostridioides difficile therapeutic. J Antimicrob Chemother 2021; 75:409-417. [PMID: 31642901 DOI: 10.1093/jac/dkz430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/12/2019] [Accepted: 09/18/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Clostridioides difficile (previously Clostridium difficile) is the leading cause of nosocomial, antibiotic-associated diarrhoea worldwide. Currently, the gold standard of treatment for C. difficile infection (CDI) is vancomycin or metronidazole, although these antibiotics also perturb the protective resident microbiota, often resulting in disease relapse. Thus, an urgent need remains for the development of new treatment strategies. Auranofin is an FDA-approved oral antirheumatic drug that was previously shown to inhibit C. difficile vegetative cell growth, toxin production and spore production in vitro. OBJECTIVES To determine the efficacy of auranofin as a CDI therapeutic by examining the effect of treatment on toxin and spore production in vitro and in vivo, and on disease outcomes in mice. METHODS C. difficile cultures were treated with auranofin and examined for effects on sporulation and toxin production by sporulation assay and ELISA, respectively. Mice were pretreated with auranofin prior to infection with C. difficile and monitored for physiological conditions, survival and gut damage compared with control animals. Faeces from mice were analysed to determine whether auranofin reduces sporulation and toxin production in vivo. RESULTS Auranofin significantly reduces sporulation and toxin production under in vitro conditions and in infected mice in vivo. Mice treated with auranofin lost less weight, displayed a significant increase in survival rates and had significantly less toxin-mediated damage in their colon and caecum compared with control mice. CONCLUSIONS Auranofin shows promise as a prospective therapeutic option for C. difficile infections.
Collapse
Affiliation(s)
- Melanie L Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Havva Pehlivanoglu
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Callum J Vidor
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Meagan L James
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Melanie J Thomson
- School of Medicine, Deakin University, Pigdons Road, Waurn Ponds, Victoria, 3216, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
21
|
Cirri D, Bartoli F, Pratesi A, Baglini E, Barresi E, Marzo T. Strategies for the Improvement of Metal-Based Chemotherapeutic Treatments. Biomedicines 2021; 9:504. [PMID: 34064364 PMCID: PMC8147839 DOI: 10.3390/biomedicines9050504] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
This article provides an overview of the various research approaches we have explored in recent years to improve metal-based agents for cancer or infection treatments. Although cisplatin, carboplatin, and oxaliplatin remain the cornerstones in tumor chemotherapy, the discovery and approval of novel inorganic anticancer drugs is a very slow process. Analogously, although a few promising inorganic drugs have found clinical application against parasitic or bacterial infections, their use remains relatively limited. Moreover, the discovery process is often affected by small therapeutic enhancements that are not attractive for the pharmaceutical industry. However, the availability of increasing mechanistic information for the modes of action of established inorganic drugs is fueling the exploration of various approaches for developing effective inorganic chemotherapy agents. Through a series of examples, some from our own research experience, we focus our attention on a number of promising strategies, including (1) drug repurposing, (2) the simple modification of the chemical structures of approved metal-based drugs, (3) testing novel drug combinations, and (4) newly synthesized complexes coupling different anticancer drugs. Accordingly, we aim to suggest and summarize a series of reliable approaches that are exploitable for the development of improved and innovative treatments.
Collapse
Affiliation(s)
- Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Francesco Bartoli
- Department of Translational Research and of New Surgical and Medical Technologies, Univerisity of Pisa, Via Risorgimento, 36, 56126 Pisa, Italy;
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), Univerisity of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy;
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy; (E.B.); (E.B.)
| |
Collapse
|
22
|
Tolbatov I, Cirri D, Marchetti L, Marrone A, Coletti C, Re N, La Mendola D, Messori L, Marzo T, Gabbiani C, Pratesi A. Mechanistic Insights Into the Anticancer Properties of the Auranofin Analog Au(PEt 3)I: A Theoretical and Experimental Study. Front Chem 2020; 8:812. [PMID: 33195032 PMCID: PMC7531625 DOI: 10.3389/fchem.2020.00812] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
Au(PEt3)I (AF-I hereafter), the iodide analog of the FDA-approved drug auranofin (AF hereafter), is a promising anticancer agent that produces its pharmacological effects through interaction with non-genomic targets such as the thioredoxin reductase system. AF-I is endowed with a very favorable biochemical profile showing potent in vitro cytotoxic activity against several cancer types including ovarian and colorectal cancer. Remarkably, in a recent publication, some of us reported that AF-I induces an almost complete and rapid remission in an orthotopic in vivo mouse model of ovarian cancer. The cytotoxic potency does not bring about highly severe side effects, making AF-I very well-tolerated even for higher doses, even more so than the pharmacologically active ones. All these promising features led us to expand our studies on the mechanistic aspects underlying the antitumor activity of AF-I. We report here on an integrated experimental and theoretical study on the reactivity of AF-I, in comparison with auranofin, toward relevant aminoacidic residues or their molecular models. Results point out that the replacement of the thiosugar moiety with iodide significantly affects the overall reactivity toward the amino acid residues histidine, cysteine, methionine, and selenocysteine. Altogether, the obtained results contribute to shed light into the enhanced antitumoral activity of AF-I compared with AF.
Collapse
Affiliation(s)
- Iogann Tolbatov
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Pisa, Italy
| | - Lorella Marchetti
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Pisa, Italy
| | - Alessandro Marrone
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Cecilia Coletti
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Nazzareno Re
- Department of Pharmacy, University "G. D'Annunzio" Chieti-Pescara, Chieti, Italy
| | | | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry "U. Schiff", University of Florence, Florence, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Pisa, Italy.,CISUP-Centro per l'Integrazione della Strumentazione Scientifica dell'Università di Pisa, University of Pisa, Pisa, Italy
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Pisa, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Ren X, Zou L, Holmgren A. Targeting Bacterial Antioxidant Systems for Antibiotics Development. Curr Med Chem 2020; 27:1922-1939. [PMID: 31589114 DOI: 10.2174/0929867326666191007163654] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 09/18/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022]
Abstract
The emergence of multidrug-resistant bacteria has become an urgent issue in modern medicine which requires novel strategies to develop antibiotics. Recent studies have supported the hypothesis that antibiotic-induced bacterial cell death is mediated by Reactive Oxygen Species (ROS). The hypothesis also highlighted the importance of antioxidant systems, the defense mechanism which contributes to antibiotic resistance. Thioredoxin and glutathione systems are the two major thiol-dependent systems which not only provide antioxidant capacity but also participate in various biological events in bacteria, such as DNA synthesis and protein folding. The biological importance makes them promising targets for novel antibiotics development. Based on the idea, ebselen and auranofin, two bacterial thioredoxin reductase inhibitors, have been found to inhibit the growth of bacteria lacking the GSH efficiently. A recent study combining ebselen and silver exhibited a strong synergistic effect against Multidrug-Resistant (MDR) Gram-negative bacteria which possess both thioredoxin and glutathione systems. These drug-repurposing studies are promising for quick clinical usage due to their well-known profile.
Collapse
Affiliation(s)
- Xiaoyuan Ren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Lili Zou
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden.,Translational Neuroscience & Neural Regeneration and Repair Institute/ Institute of Cell Therapy, The First Hospital of Yichang, Three Gorges University, 443000 Yichang, China
| | - Arne Holmgren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
24
|
Martín-Encinas E, Conejo-Rodríguez V, Miguel JA, Martínez-Ilarduya JM, Rubiales G, Knudsen BR, Palacios F, Alonso C. Novel phosphine sulphide gold(i) complexes: topoisomerase I inhibitors and antiproliferative agents. Dalton Trans 2020; 49:7852-7861. [DOI: 10.1039/d0dt01467b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gold(i) increases the cytotoxicity of phosphine sulfide quinolines against cancer cell lines, while heterocycles maintain the TopI inhibitory activity.
Collapse
Affiliation(s)
- Endika Martín-Encinas
- Department of Organic Chemistry I
- Faculty of Pharmacy
- University of Basque Country (UPV/EHU)
- Vitoria-Gasteiz
- Spain
| | | | - Jesús A. Miguel
- IU CINQUIMA/Química Inorgánica
- Faculty of Science
- University of Valladolid
- Valladolid
- Spain
| | | | - Gloria Rubiales
- Department of Organic Chemistry I
- Faculty of Pharmacy
- University of Basque Country (UPV/EHU)
- Vitoria-Gasteiz
- Spain
| | - Birgitta R. Knudsen
- Department of Molecular Biology and Genetics and Interdisciplinary Nanoscience Center (iNANO)
- University of Aarhus
- Aarhus
- Denmark
| | - Francisco Palacios
- Department of Organic Chemistry I
- Faculty of Pharmacy
- University of Basque Country (UPV/EHU)
- Vitoria-Gasteiz
- Spain
| | - Concepción Alonso
- Department of Organic Chemistry I
- Faculty of Pharmacy
- University of Basque Country (UPV/EHU)
- Vitoria-Gasteiz
- Spain
| |
Collapse
|
25
|
She P, Liu Y, Wang Y, Tan F, Luo Z, Wu Y. Antibiofilm efficacy of the gold compound auranofin on dual species biofilms of
Staphylococcus aureus
and
Candida
sp. J Appl Microbiol 2019; 128:88-101. [PMID: 31509623 DOI: 10.1111/jam.14443] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/12/2019] [Accepted: 08/31/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yiqing Liu
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yangxia Wang
- Department of Clinical Laboratory The First Affiliated Hospital of Zhengzhou University Zhengzhou P.R. China
| | - Fang Tan
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Zhen Luo
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| | - Yong Wu
- Department of Clinical Laboratory The Third Xiangya Hospital of Central South University Changsha P.R. China
| |
Collapse
|
26
|
She P, Zhou L, Li S, Liu Y, Xu L, Chen L, Luo Z, Wu Y. Synergistic Microbicidal Effect of Auranofin and Antibiotics Against Planktonic and Biofilm-Encased S. aureus and E. faecalis. Front Microbiol 2019; 10:2453. [PMID: 31708908 PMCID: PMC6821689 DOI: 10.3389/fmicb.2019.02453] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022] Open
Abstract
Methicillin-resistant/susceptible Staphylococcus aureus (MRSA/MSSA) and Enterococcus faecalis strains are often found in community- and hospital-acquired infections. The single use of conventional antibiotics hardly completely kills the bacterial cells of interest, especially in the form of biofilms. Thus, drug repurposing and antimicrobial combination are promising ways to solve this problem. Antimicrobial susceptibility assays against cocci in a suspension and in a biofilm mode of growth were performed with broth microdilution methods. Checkerboard assays and the cutaneous mouse infection model were used to examine the activity of auranofin and conventional antibiotics alone and in combination. In the present study, auranofin possesses potent antimicrobial activities against both planktonic cells and biofilms with minimum inhibitory concentrations ranging 0.125–0.5 mg/L. Auranofin in combination with linezolid or fosfomycin showed synergistic antimicrobial activities against S. aureus MSSA and MRSA both in vitro and in vivo. Similarly, auranofin also behaved synergistic effect with chloramphenicol against E. faecalis. Additionally, auranofin improved the antibiofilm efficacy of chloramphenicol and linezolid, even on the biofilms grown on a catheter surface. Though, S. epidermidis showed significant susceptibility to AF treatment, no synergistic antimicrobial effects were observed with antibiotics we tested. In all, the use of a combination of auranofin with linezolid, fosfomycin, and chloramphenicol can provide a synergistic microbicidal effect in vitro and in vivo, which rapidly enhances antimicrobial activity and may help prevent or delay the emergence of resistance.
Collapse
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Linying Zhou
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shijia Li
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yiqing Liu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lanlan Xu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
27
|
Epstein TD, Wu B, Moulton KD, Yan M, Dube DH. Sugar-Modified Analogs of Auranofin Are Potent Inhibitors of the Gastric Pathogen Helicobacter pylori. ACS Infect Dis 2019; 5:1682-1687. [PMID: 31487153 PMCID: PMC7123778 DOI: 10.1021/acsinfecdis.9b00251] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori (H. pylori) infection poses a worldwide public health crisis, as chronic infection is rampant and can lead to gastric ulcers, gastritis, and gastric cancer. Unfortunately, frontline therapies cause harmful side effects and are often ineffective due to antibiotic resistance. The FDA-approved drug auranofin is a gold complex with a Au(I) core coordinated with triethylphosphine and peracetylated thioglucose as the ligands. Auranofin is used for the treatment of rheumatoid arthritis and also displays potent activity against H. pylori. One of auranofin's modes of action involves cell death by disrupting cellular thiol-redox balance maintained by thioredoxin reductase (TrxR), but this disruption leads to unwanted side effects due to mammalian cell toxicity. Here, we developed and tested sugar-modified analogs of auranofin as potential antibiotics against H. pylori, with the rationale that modulating the sugar moiety would bias uptake by targeting bacterial cells and mitigating mammalian cell toxicity. Sugar-modified auranofin analogs displayed micromolar minimum inhibitory concentrations against H. pylori, maintained nanomolar inhibitory activity against the target enzyme TrxR, and caused reduced toxicity to mammalian cells. Taken together, our results suggest that structurally modifying the sugar component of auranofin has the potential to yield superior antibiotics for the treatment of H. pylori infection. Broadly, glyco-tailoring is an attractive approach for repurposing approved drugs.
Collapse
Affiliation(s)
- Tessa D. Epstein
- Department of Chemistry & Biochemistry, Bowdoin College, 6600 College Station, Brunswick, ME 04011, USA
| | - Bin Wu
- Department of Chemistry, University of Massachusetts Lowell, 1 University Ave., Lowell, MA 01854, USA
| | - Karen D. Moulton
- Department of Chemistry & Biochemistry, Bowdoin College, 6600 College Station, Brunswick, ME 04011, USA
| | - Mingdi Yan
- Department of Chemistry, University of Massachusetts Lowell, 1 University Ave., Lowell, MA 01854, USA
| | - Danielle H. Dube
- Department of Chemistry & Biochemistry, Bowdoin College, 6600 College Station, Brunswick, ME 04011, USA
| |
Collapse
|
28
|
Wu B, Yang X, Yan M. Synthesis and Structure-Activity Relationship Study of Antimicrobial Auranofin against ESKAPE Pathogens. J Med Chem 2019; 62:7751-7768. [PMID: 31386365 PMCID: PMC7941214 DOI: 10.1021/acs.jmedchem.9b00550] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Auranofin, an FDA-approved arthritis drug, has recently been repurposed as a potential antimicrobial agent; it performed well against many Gram-positive bacteria, including multidrug resistant strains. It is, however, inactive toward Gram-negative bacteria, for which we are in dire need of new therapies. In this work, 40 auranofin analogues were synthesized by varying the structures of the thiol and phosphine ligands, and their activities were tested against ESKAPE pathogens. The study identified compounds that exhibited bacterial inhibition (MIC) and killing (MBC) activities up to 65 folds higher than that of auranofin, making them effective against Gram-negative pathogens. Both thiol and the phosphine structures influence the activities of the analogues. The trimethylphosphine and triethylphosphine ligands gave the highest activities against Gram-negative and Gram-positive bacteria, respectively. Our SAR study revealed that the thiol ligand is also very important, the structure of which can modulate the activities of the AuI complexes for both Gram-negative and Gram-positive bacteria. Moreover, these analogues had mammalian cell toxicities either similar to or lower than that of auranofin.
Collapse
Affiliation(s)
- Bin Wu
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Xiaojian Yang
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mingdi Yan
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| |
Collapse
|
29
|
Gold(I/III)-Phosphine Complexes as Potent Antiproliferative Agents. Sci Rep 2019; 9:12335. [PMID: 31451718 PMCID: PMC6710276 DOI: 10.1038/s41598-019-48584-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/07/2019] [Indexed: 11/23/2022] Open
Abstract
The reaction of gold reagents [HAuCl4•3H2O], [AuCl(tht)], or cyclometalated gold(III) precursor, [C^NAuCl2] with chiral ((R,R)-(-)-2,3-bis(t-butylmethylphosphino) quinoxaline) and non-chiral phosphine (1,2-Bis(diphenylphosphino)ethane, dppe) ligands lead to distorted Au(I), (1, 2, 4, 5) and novel cyclometalated Au(III) complexes (3, 6). These gold compounds were characterized by multinuclear NMR, microanalysis, mass spectrometry, and X-ray crystallography. The inherent electrochemical properties of the gold complexes were also studied by cyclic voltammetry and theoretical insight of the complexes was gained by density functional theory and TD-DFT calculations. The complexes effectively kill cancer cells with IC50 in the range of ~0.10–2.53 μΜ across K562, H460, and OVCAR8 cell lines. In addition, the retinal pigment epithelial cell line, RPE-Neo was used as a healthy cell line for comparison. Differential cellular uptake in cancer cells was observed for the compounds by measuring the intracellular accumulation of gold using ICP-OES. Furthermore, the compounds trigger early – late stage apoptosis through potential disruption of redox homeostasis. Complexes 1 and 3 induce predominant G1 cell cycle arrest. Results presented in this report suggest that stable gold-phosphine complexes with variable oxidation states hold promise in anticancer drug discovery and need further development.
Collapse
|
30
|
Auranofin is an effective agent against clinical isolates of Staphylococcus aureus. Future Med Chem 2019; 11:1417-1425. [PMID: 31298580 DOI: 10.4155/fmc-2018-0544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: The orphan drug auranofin was recently found to exhibit antimicrobial properties. Materials & methods: We explored the efficacy of auranofin by evaluating the minimal inhibitory concentration against a collection of over 500 clinical isolates derived from multiple institutions, inclusive of drug resistant strains. Our evaluation also included continuous exposure of bacteria to auranofin. Results & conclusion: We found that minimal inhibitory concentrations ranged between 0.125 and 1 mg/l, exerting robust antimicrobial activity against a sizeable clinical collection of the bacteria. Further, we evaluated the propensity of the methicillin-resistant Staphylococcus aureus strain MW2 to develop resistance through extended exposure to auranofin. After 25 days, the bacteria remained susceptible. Our data suggest that resistance mechanisms do not currently exist to block auranofin antimicrobial activity.
Collapse
|
31
|
Jang H, Eom Y. Repurposing auranofin to combat uropathogenic
Escherichia coli
biofilms. J Appl Microbiol 2019; 127:459-471. [DOI: 10.1111/jam.14312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/27/2019] [Accepted: 05/10/2019] [Indexed: 12/29/2022]
Affiliation(s)
- H.‐I. Jang
- Department of Medical Sciences, College of Medical Sciences Soonchunhyang University Asan Republic of Korea
| | - Y.‐B. Eom
- Department of Medical Sciences, College of Medical Sciences Soonchunhyang University Asan Republic of Korea
- Department of Biomedical Laboratory Science, College of Medical Sciences Soonchunhyang University Asan Republic of Korea
| |
Collapse
|
32
|
Marzo T, Massai L, Pratesi A, Stefanini M, Cirri D, Magherini F, Becatti M, Landini I, Nobili S, Mini E, Crociani O, Arcangeli A, Pillozzi S, Gamberi T, Messori L. Replacement of the Thiosugar of Auranofin with Iodide Enhances the Anticancer Potency in a Mouse Model of Ovarian Cancer. ACS Med Chem Lett 2019; 10:656-660. [PMID: 30996813 DOI: 10.1021/acsmedchemlett.9b00007] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years, a few successful attempts were made to repurpose the clinically approved antiarthritic gold drug, Auranofin (AF), as an anticancer agent. The present study shows that the iodido(triethylphosphine)gold(I) complex, (Et 3 PAuI hereafter)-an AF analogue where the thiosugar ligand is simply replaced by one iodide ligand-manifests a solution chemistry resembling that of AF and exerts similar cytotoxic and proapoptotic effects on A2780 human ovarian cancer cells in vitro. However, when evaluated in a preclinical orthotopic model of ovarian cancer, Et 3 PAuI produces a far superior anticancer action than AF inducing a nearly complete tumor remission. The highly promising in vivo performances here documented for Et 3 PAuI warrant its further evaluation as a drug candidate for ovarian cancer treatment.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Lara Massai
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Alessandro Pratesi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Matteo Stefanini
- DI.V.A.L. Toscana s.r.l., via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| | - Damiano Cirri
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Francesca Magherini
- Department of Biochemical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Matteo Becatti
- Department of Biochemical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, viale Pieraccini 6, 50139 Firenze, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, viale Pieraccini 6, 50139 Firenze, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, viale Pieraccini 6, 50139 Firenze, Italy
| | - Olivia Crociani
- DI.V.A.L. Toscana s.r.l., via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Serena Pillozzi
- DI.V.A.L. Toscana s.r.l., via Madonna del Piano 6, 50019 Sesto Fiorentino, Italy
- Department of Experimental and Clinical Medicine, University of Florence, viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Tania Gamberi
- Department of Biochemical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, viale G.B. Morgagni 50, 50134 Firenze, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
33
|
Mazzei L, Wenzel MN, Cianci M, Palombo M, Casini A, Ciurli S. Inhibition Mechanism of Urease by Au(III) Compounds Unveiled by X-ray Diffraction Analysis. ACS Med Chem Lett 2019; 10:564-570. [PMID: 30996797 DOI: 10.1021/acsmedchemlett.8b00585] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/04/2019] [Indexed: 11/29/2022] Open
Abstract
The nickel-dependent enzyme urease is a virulence factor for a large number of critical human pathogens, making this enzyme a potential target of therapeutics for the treatment of resistant bacterial infections. In the search for novel urease inhibitors, five selected coordination and organometallic Au(III) compounds containing N∧N or C∧N and C∧N∧N ligands were tested for their inhibitory effects against Canavalia ensiformis (jack bean) urease. The results showed potent inhibition effects with IC50 values in the nanomolar range. The 2.14 Å resolution crystal structure of Sporosarcina pasteurii urease inhibited by the most effective Au(III) compound [Au(PbImMe)Cl2]PF6 (PbImMe = 1-methyl-2-(pyridin-2-yl)-benzimidazole) reveals the presence of two Au ions bound to the conserved triad αCys322/αHis323/αMet367. The binding of the Au ions to these residues blocks the movement of a flap, located at the edge of the active site channel and essential for enzyme catalysis, completely obliterating the catalytic activity of urease. Overall, the obtained results constitute the basis for the design of new gold complexes as selective urease inhibitors with future antibacterial applications.
Collapse
Affiliation(s)
- Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| | - Margot N. Wenzel
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, I-60131 Ancona, Italy
| | - Marta Palombo
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| | - Angela Casini
- School of Chemistry, Cardiff University, Main Building, Park Place, CF10 3AT Cardiff, United Kingdom
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Viale Giuseppe Fanin 40, I-40127 Bologna, Italy
| |
Collapse
|
34
|
Liu H, Shukla S, Vera-González N, Tharmalingam N, Mylonakis E, Fuchs BB, Shukla A. Auranofin Releasing Antibacterial and Antibiofilm Polyurethane Intravascular Catheter Coatings. Front Cell Infect Microbiol 2019; 9:37. [PMID: 30873389 PMCID: PMC6403144 DOI: 10.3389/fcimb.2019.00037] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Intravascular catheter related bloodstream infections (CRBSIs) are a leading cause of hospital-acquired infections worldwide, resulting not only in the burden of cost and morbidity for patients but also in the over-consumption of medical resources for hospitals and health care organizations. In this study, a novel auranofin releasing antibacterial and antibiofilm polyurethane (PU) catheter coating was developed and investigated for future use in preventing CRBSIs. Auranofin is an antirheumatic drug with recently identified antimicrobial properties. The drug carrier, PU, acts as a barrier surrounding the antibacterial agent, auranofin, to extend the drug release profile and improve its long-term antibacterial and antibiofilm efficacy and potentially the length of catheter implantation within a patient. The PU+auranofin coatings developed here were found to be highly stretchable (exhibiting ~500% percent elongation), which is important for the compliance of the material on a flexible catheter. PU+auranofin coated catheters were able to inhibit the growth of methicillin-resistant Staphylococcus aureus (MRSA) for 8 to 26 days depending on the specific drug concentration utilized during the dip coating process. The PU+auranofin coated catheters were also able to completely inhibit MRSA biofilm formation in vitro, an effect that was not observed with auranofin or PU alone. Lastly, these coatings were found to be hemocompatible with human erythrocytes and maintain liver cell viability.
Collapse
Affiliation(s)
- Hanyang Liu
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Shashank Shukla
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Noel Vera-González
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| | - Nagendran Tharmalingam
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School and Brown University, Providence, RI, United States
| | - Anita Shukla
- Center for Biomedical Engineering, School of Engineering, Institute for Molecular and Nanoscale Innovation, Brown University, Providence, RI, United States
| |
Collapse
|
35
|
Jang HJ, Chung IY, Lim C, Chung S, Kim BO, Kim ES, Kim SH, Cho YH. Redirecting an Anticancer to an Antibacterial Hit Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:350. [PMID: 30858845 PMCID: PMC6398426 DOI: 10.3389/fmicb.2019.00350] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/11/2019] [Indexed: 01/18/2023] Open
Abstract
YM155 is a clinically evaluated anticancer with a fused naphthoquinone-imidazolium scaffold. In this study, we demonstrated that based on weak or cryptic antibacterial activity of YM155 against methicillin-resistant Staphylococcus aureus (MRSA) (MIC of 50 μg/ml), some congeneric compounds with short alkyl chains (e.g., c5 with a hexyl chain) at the N3 position of the scaffold, displayed more potent antibacterial activity against MRSA (MIC of 3.13 μg/ml), which is in a clinically achievable range. Their antibacterial activity was evident against Gram-negative bacteria, only in the presence of the outer membrane-permeabilizing agent, polymyxin B. The antibacterial efficacy of c5 was confirmed using the Drosophila systemic infection model. We also characterized five spontaneous c5-resistant MRSA mutants that carry mutations in the ubiE gene, for quinone metabolism and respiratory electron transfer, and subsequently exhibited reduced respiration activity. The antibacterial activity of c5 was compromised either by an antioxidant, N-acetylcysteine, or in an anaerobic condition. These suggest that the antibacterial mechanism of c5 involves the generation of reactive oxygen species (ROS), presumably during respiratory electron transport. This study provides an insight into "drug redirecting," through a chemical modification, based on an ROS-generating pharmacophore.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - In-Young Chung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Changjin Lim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Sungkyun Chung
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Bi-O Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Eun Sook Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Seok-Ho Kim
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| |
Collapse
|
36
|
Marzo T, Cirri D, Pollini S, Prato M, Fallani S, Cassetta MI, Novelli A, Rossolini GM, Messori L. Auranofin and its Analogues Show Potent Antimicrobial Activity against Multidrug-Resistant Pathogens: Structure-Activity Relationships. ChemMedChem 2018; 13:2448-2454. [DOI: 10.1002/cmdc.201800498] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/24/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Tiziano Marzo
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”; University of Florence; Via della Lastruccia 3 50019 Sesto Fiorentino Italy
- Department of Chemistry and Industrial Chemistry (DCCI); University of Pisa; Via Moruzzi 13 56124 Pisa Italy
| | - Damiano Cirri
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”; University of Florence; Via della Lastruccia 3 50019 Sesto Fiorentino Italy
| | - Simona Pollini
- Department of Experimental and Clinical Medicine (DMSC); University of Florence; Largo Brambilla 3 50134 Firenze Italy
- Microbiology and Virology Unit; Azienda Ospedaliero-Universitaria Careggi (AOUC); Largo Brambilla 3 50134 Firenze Italy
| | - Marco Prato
- Department of Experimental and Clinical Medicine (DMSC); University of Florence; Largo Brambilla 3 50134 Firenze Italy
| | - Stefania Fallani
- Department of Health Sciences (DSS); University of Florence; Viale Pieraccini 6 50139 Florence Italy
| | - Maria Iris Cassetta
- Department of Health Sciences (DSS); University of Florence; Viale Pieraccini 6 50139 Florence Italy
| | - Andrea Novelli
- Department of Health Sciences (DSS); University of Florence; Viale Pieraccini 6 50139 Florence Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine (DMSC); University of Florence; Largo Brambilla 3 50134 Firenze Italy
- Microbiology and Virology Unit; Azienda Ospedaliero-Universitaria Careggi (AOUC); Largo Brambilla 3 50134 Firenze Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”; University of Florence; Via della Lastruccia 3 50019 Sesto Fiorentino Italy
| |
Collapse
|
37
|
Torres NS, Montelongo-Jauregui D, Abercrombie JJ, Srinivasan A, Lopez-Ribot JL, Ramasubramanian AK, Leung KP. Antimicrobial and Antibiofilm Activity of Synergistic Combinations of a Commercially Available Small Compound Library With Colistin Against Pseudomonas aeruginosa. Front Microbiol 2018; 9:2541. [PMID: 30410476 PMCID: PMC6209680 DOI: 10.3389/fmicb.2018.02541] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/04/2018] [Indexed: 01/21/2023] Open
Abstract
Biofilm-associated Pseudomonas aeruginosa infections remain a significant clinical challenge since the conventional antibiotic treatment or combination therapies are largely ineffective; and new approaches are needed. To circumvent the major challenges associated with discovery of new antimicrobials, we have screened a library of compounds that are commercially available and approved by the FDA (Prestwick Chemical Library) against P. aeruginosa for effective antimicrobial and anti-biofilm activity. A preliminary screen of the Prestwick Chemical Library alone did not yield any repositionable candidates, but in a screen of combinations with a fixed sub-inhibitory concentration of the antibiotic colistin we observed 10 drugs whose bacterial inhibiting activity was reproducibly enhanced, seven of which were enhanced by more than 50%. We performed checkerboard assays of these seven drugs in combination with colistin against planktonic cells, and analysis of their interactions over the complete combination matrix using the Zero Interaction Potency (ZIP) model revealed interactions that varied from highly synergistic to completely antagonistic. Of these, five combinations that showed synergism were down-selected and tested against preformed biofilms of P. aeruginosa. Two of the five combinations were active against preformed biofilms of both laboratory and clinical strain of P. aeruginosa, resulting in a 2-log reduction in culturable cells. In summary, we have identified synergistic combinations of five commercially available, FDA-approved drugs and colistin that show antimicrobial activity against planktonic P. aeruginosa (Clomiphene Citrate, Mitoxantrone Dihydrochloride, Methyl Benzethonium Chloride, Benzethonium Chloride, and Auranofin) as well as two combinations (Auranofin and Clomiphene Citrate) with colistin that show antibiofilm activity.
Collapse
Affiliation(s)
- Nelson S Torres
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States.,Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Daniel Montelongo-Jauregui
- Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Johnathan J Abercrombie
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States
| | - Anand Srinivasan
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, United States.,Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jose L Lopez-Ribot
- Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Anand K Ramasubramanian
- Department of Chemical and Materials Engineering, San José State University, San José, CA, United States
| | - Kai P Leung
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States
| |
Collapse
|
38
|
Warżajtis B, Glišić BĐ, Savić ND, Pavic A, Vojnovic S, Veselinović A, Nikodinovic-Runic J, Rychlewska U, Djuran MI. Mononuclear gold(iii) complexes with l-histidine-containing dipeptides: tuning the structural and biological properties by variation of the N-terminal amino acid and counter anion. Dalton Trans 2018; 46:2594-2608. [PMID: 28155927 DOI: 10.1039/c6dt04862e] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gold(iii) complexes with different l-histidine-containing dipeptides, [Au(Gly-l-His-NA,NP,N3)Cl]Cl·3H2O (1a), [Au(Gly-l-His-NA,NP,N3)Cl]NO3·1.25H2O (1b), [Au(l-Ala-l-His-NA,NP,N3)Cl][AuCl4]·H2O (2a), [Au(l-Ala-l-His-NA,NP,N3)Cl]NO3·2.5H2O (2b), [Au(l-Val-l-His-NA,NP,N3)Cl]Cl·2H2O (3), [Au(l-Leu-l-His-NA,NP,N3)Cl]Cl (4a) and [Au(l-Leu-l-His-NA,NP,N3)Cl][AuCl4]·H2O (4b), have been synthesized and structurally characterized by spectroscopic (1H NMR, IR and UV-vis) and single-crystal X-ray diffraction techniques. The antimicrobial efficiency of these gold(iii) complexes, along with K[AuCl4] and the corresponding dipeptides, was evaluated against the broad panel of Gram-positive and Gram-negative bacteria and fungi, displaying their moderate inhibiting activity. Moreover, the cytotoxic properties of the investigated complexes were assessed against the normal human lung fibroblast cell line (MRC5) and two human cancer, cervix (HeLa) and lung (A549) cell lines. None of the complexes exerted significant cytotoxic activity; nevertheless complexes that did show selectivity in terms of cancer vs. normal cell lines (2a/b and 4a/b) have been evaluated using zebrafish (Danio rerio) embryos for toxicity and antiangiogenic potential. Although the gold(iii) complexes achieved an antiangiogenic effect comparable to the known angiogenic inhibitors auranofin and sunitinib malate at 30-fold higher concentrations, they had no cardiovascular side effects, which commonly accompany auranofin and sunitinib malate treatment. Finally, binding of the gold(iii) complexes to the active sites of both human and bacterial (Escherichia coli) thioredoxin reductases (TrxRs) was demonstrated by conducting a molecular docking study, suggesting that the mechanism of biological action of these complexes can be associated with their interaction with the TrxR active site.
Collapse
Affiliation(s)
- Beata Warżajtis
- Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89B, 61-614 Poznań, Poland.
| | - Biljana Đ Glišić
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia.
| | - Nada D Savić
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia.
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia.
| | - Sandra Vojnovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia.
| | | | - Jasmina Nikodinovic-Runic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia.
| | - Urszula Rychlewska
- Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89B, 61-614 Poznań, Poland.
| | - Miloš I Djuran
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia.
| |
Collapse
|
39
|
AbdelKhalek A, Abutaleb NS, Elmagarmid KA, Seleem MN. Repurposing auranofin as an intestinal decolonizing agent for vancomycin-resistant enterococci. Sci Rep 2018; 8:8353. [PMID: 29844350 PMCID: PMC5974018 DOI: 10.1038/s41598-018-26674-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/10/2018] [Indexed: 02/07/2023] Open
Abstract
Multidrug-resistant enterococcal pathogens, especially vancomycin-resistant enterococci (VRE), are among the pathogens that require new antibiotic innovation. The colonization of the gut represents a major pathway by which VRE can cause infection and spread to other patients. In the current study, auranofin (FDA-approved rheumatoid arthritis drug) is evaluated for its potential use as a decolonizing agent for VRE. Auranofin was found to exert potent antimicrobial activity against a wide range of enterococcal clinical isolates with a minimum inhibitory concentration of 1 μg/mL. No resistant mutants could be developed against auranofin over the course of 14 passages. Auranofin was also found to exert potent anti-biofilm activity against VRE. Auranofin was superior to linezolid, the drug of choice for VRE infection treatment, in the in vivo mouse model. Auranofin significantly reduced the VRE burden in feces, cecum, and ileum contents after 8 days of treatment. Accordingly, this study provides valuable evidence that auranofin has significant promise as a novel gastrointestinal decolonizing agent for VRE.
Collapse
Affiliation(s)
- Ahmed AbdelKhalek
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Nader S Abutaleb
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Khalifa A Elmagarmid
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, IN, 47907, USA.
| |
Collapse
|
40
|
May HC, Yu JJ, Guentzel MN, Chambers JP, Cap AP, Arulanandam BP. Repurposing Auranofin, Ebselen, and PX-12 as Antimicrobial Agents Targeting the Thioredoxin System. Front Microbiol 2018; 9:336. [PMID: 29556223 PMCID: PMC5844926 DOI: 10.3389/fmicb.2018.00336] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 02/12/2018] [Indexed: 01/23/2023] Open
Abstract
As microbial resistance to drugs continues to rise at an alarming rate, finding new ways to combat pathogens is an issue of utmost importance. Development of novel and specific antimicrobial drugs is a time-consuming and expensive process. However, the re-purposing of previously tested and/or approved drugs could be a feasible way to circumvent this long and costly process. In this review, we evaluate the U.S. Food and Drug Administration tested drugs auranofin, ebselen, and PX-12 as antimicrobial agents targeting the thioredoxin system. These drugs have been shown to act on bacterial, fungal, protozoan, and helminth pathogens without significant toxicity to the host. We propose that the thioredoxin system could serve as a useful therapeutic target with broad spectrum antimicrobial activity.
Collapse
Affiliation(s)
- Holly C. May
- South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, TX, United States
- Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, United States
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, TX, United States
- Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, United States
| | - M. N. Guentzel
- South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, TX, United States
- Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, United States
| | - James P. Chambers
- South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, TX, United States
- Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, United States
| | - Andrew P. Cap
- United States Army Institute for Surgical Research, San Antonio Military Medical Center, San Antonio, TX, United States
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, TX, United States
- Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
41
|
Liao X, Yang F, Li H, So PK, Yao Z, Xia W, Sun H. Targeting the Thioredoxin Reductase-Thioredoxin System from Staphylococcus aureus by Silver Ions. Inorg Chem 2017; 56:14823-14830. [PMID: 29182243 DOI: 10.1021/acs.inorgchem.7b01904] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The thioredoxin system, which is composed of NADPH, thioredoxin reductase (TrxR), and thioredoxin (Trx), is one of the major disulfide reductase systems used by bacteria against oxidative stress. In particular, this reductase system is crucial for the survival of the pathogenic bacterium Staphylococcus aureus, which lacks a natural glutathione/glutaredoxin (Grx) system. Although silver ions and silver-containing materials have been used as antibacterial agents for centuries, the antibacterial mechanism of silver is not well-understood. Herein, we demonstrate that silver ions bind to the active sites of S. aureus TrxR and Trx with dissociation constants of 1.4 ± 0.1 μM and 15.0 ± 5.0 μM and stoichiometries of 1 and 2 Ag+ ions per protein, respectively. Importantly, silver ion binding leads to oligomerization and functional disruption of TrxR as well as Trx. Silver also depleted intracellular thiol levels in S. aureus, disrupting bacterial thiol-redox homeostasis. Our study provides new insights into the antibacterial mechanism of silver ions. Moreover, the Trx and TrxR system might serve as a feasible target for the design of antibacterial drugs.
Collapse
Affiliation(s)
- Xiangwen Liao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University , Guangzhou, China , 510275.,Hunan Provincial Key Laboratory for Ethnic Dong Medicine Research, Hunan University of Medicine , Huaihua, China , 418000
| | - Fang Yang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University , Guangzhou, China , 510275
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong , Pokfulam Road, Hong Kong SAR, China
| | - Pui-Kin So
- State Key Laboratory for Chirosciences and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University , Hung Hom, Kowloon, Hong Kong SAR, China
| | - Zhongping Yao
- State Key Laboratory for Chirosciences and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University , Hung Hom, Kowloon, Hong Kong SAR, China
| | - Wei Xia
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University , Guangzhou, China , 510275
| | - Hongzhe Sun
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University , Guangzhou, China , 510275.,Department of Chemistry, The University of Hong Kong , Pokfulam Road, Hong Kong SAR, China
| |
Collapse
|
42
|
Marzo T, Cirri D, Gabbiani C, Gamberi T, Magherini F, Pratesi A, Guerri A, Biver T, Binacchi F, Stefanini M, Arcangeli A, Messori L. Auranofin, Et 3PAuCl, and Et 3PAuI Are Highly Cytotoxic on Colorectal Cancer Cells: A Chemical and Biological Study. ACS Med Chem Lett 2017; 8:997-1001. [PMID: 29057040 DOI: 10.1021/acsmedchemlett.7b00162] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 09/06/2017] [Indexed: 12/23/2022] Open
Abstract
The solution behavior of auranofin, Et3PAuCl and Et3PAuI, as well as their interactions with hen egg white lysozyme, single strand oligonucleotide, and ds-DNA were comparatively analyzed through NMR spectroscopy, ESI-MS, ethidium bromide displacement, DNA melting and viscometric tests. The cytotoxic effects toward representative colorectal cancer cell lines were found to be strong and similar in the three cases and a good correlation could be established between the cytotoxicity and the ability to inhibit thioredoxin reductase; remarkably, in vivo acute toxicity experiments for Et3PAuI confirmed that, similarly to auranofin, this drug is well tolerated in a murine model. Overall, a very similar profile emerges for Et3PAuI and Et3PAuCl, which retain the potent cytotoxic effects of auranofin while showing some peculiar features. These results demonstrate that the presence of the thiosugar moiety is not mandatory for the pharmacological action, suggesting that the tuning of some relevant chemical properties such as lipophilicity could be exploited to improve bioavailability, with no loss of the pharmacological effects.
Collapse
Affiliation(s)
- Tiziano Marzo
- Department
of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Moruzzi, 13, 56124 Pisa, Italy
- Laboratory
of Metals in Medicine (MetMed), Department of Chemistry “U.
Schiff”, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Damiano Cirri
- Laboratory
of Metals in Medicine (MetMed), Department of Chemistry “U.
Schiff”, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Chiara Gabbiani
- Department
of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Moruzzi, 13, 56124 Pisa, Italy
| | - Tania Gamberi
- Department
of Biochemical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy
| | - Francesca Magherini
- Department
of Biochemical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy
| | - Alessandro Pratesi
- Laboratory
of Metals in Medicine (MetMed), Department of Chemistry “U.
Schiff”, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Annalisa Guerri
- Laboratory
of Metals in Medicine (MetMed), Department of Chemistry “U.
Schiff”, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Tarita Biver
- Department
of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Moruzzi, 13, 56124 Pisa, Italy
| | - Francesca Binacchi
- Department
of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Moruzzi, 13, 56124 Pisa, Italy
| | - Matteo Stefanini
- DI.V.A.L Toscana S.R.L., Via Madonna
del Piano, 6, 50019 Sesto Fiorentino, Italy
| | - Annarosa Arcangeli
- Department
of Experimental and Clinical Medicine, University of Florence, Viale GB
Morgagni 50, 50134 Firenze, Italy
| | - Luigi Messori
- Laboratory
of Metals in Medicine (MetMed), Department of Chemistry “U.
Schiff”, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
43
|
Schmidt C, Karge B, Misgeld R, Prokop A, Brönstrup M, Ott I. Biscarbene gold(i) complexes: structure-activity-relationships regarding antibacterial effects, cytotoxicity, TrxR inhibition and cellular bioavailability. MEDCHEMCOMM 2017; 8:1681-1689. [PMID: 30108879 PMCID: PMC6072206 DOI: 10.1039/c7md00269f] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/20/2017] [Indexed: 12/25/2022]
Abstract
A series of gold(i) complexes with two N-heterocyclic carbene ligands (biscarbene gold complexes) were prepared and evaluated for their effects against cancer cells and pathogenic bacteria. Proliferation inhibition was observed in cancer cells and in Gram-positive bacteria, whereas Gram-negative bacteria were less sensitive towards the compounds. The protein binding and cellular uptake were quantified and the combined results indicated a strong correlation between cellular bioavailability and antiproliferative effects. The biscarbene gold complexes inhibited bacterial and mammalian TrxRs with low to moderate potency. However, based on the obtained structure-activity-relationships and the high cellular accumulation levels, TrxR inhibition can be considered as a relevant contributor to the cellular pharmacology of biscarbene gold(i) complexes.
Collapse
Affiliation(s)
- Claudia Schmidt
- Institute of Medicinal and Pharmaceutical Chemistry , Technische Universität Braunschweig , Beethovenstr. 55 , 38106 Braunschweig , Germany .
| | - Bianka Karge
- Department of Chemical Biology , Helmholtz Centre for Infection Research and German Centre for Infection Research (DZIF) , Inhoffenstr. 7 , 38124 Braunschweig , Germany
| | - Rainer Misgeld
- Department of Pediatric Oncology , Children's Hospital Cologne , Amsterdamer Strasse 59 , 50735 Cologne , Germany
| | - Aram Prokop
- Department of Pediatric Oncology , Children's Hospital Cologne , Amsterdamer Strasse 59 , 50735 Cologne , Germany
| | - Mark Brönstrup
- Department of Chemical Biology , Helmholtz Centre for Infection Research and German Centre for Infection Research (DZIF) , Inhoffenstr. 7 , 38124 Braunschweig , Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry , Technische Universität Braunschweig , Beethovenstr. 55 , 38106 Braunschweig , Germany .
| |
Collapse
|
44
|
Thangamani S, Maland M, Mohammad H, Pascuzzi PE, Avramova L, Koehler CM, Hazbun TR, Seleem MN. Repurposing Approach Identifies Auranofin with Broad Spectrum Antifungal Activity That Targets Mia40-Erv1 Pathway. Front Cell Infect Microbiol 2017; 7:4. [PMID: 28149831 PMCID: PMC5241286 DOI: 10.3389/fcimb.2017.00004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022] Open
Abstract
Current antifungal therapies have limited effectiveness in treating invasive fungal infections. Furthermore, the development of new antifungal is currently unable to keep pace with the urgent demand for safe and effective new drugs. Auranofin, an FDA-approved drug for the treatment of rheumatoid arthritis, inhibits growth of a diverse array of clinical isolates of fungi and represents a new antifungal agent with a previously unexploited mechanism of action. In addition to auranofin's potent antifungal activity against planktonic fungi, this drug significantly reduces the metabolic activity of Candida cells encased in a biofilm. Unbiased chemogenomic profiling, using heterozygous S. cerevisiae deletion strains, combined with growth assays revealed three probable targets for auranofin's antifungal activity—mia40, acn9, and coa4. Mia40 is of particular interest given its essential role in oxidation of cysteine rich proteins imported into the mitochondria. Biochemical analysis confirmed auranofin targets the Mia40-Erv1 pathway as the drug inhibited Mia40 from interacting with its substrate, Cmc1, in a dose-dependent manner similar to the control, MB-7. Furthermore, yeast mitochondria overexpressing Erv1 were shown to exhibit resistance to auranofin as an increase in Cmc1 import was observed compared to wild-type yeast. Further in vivo antifungal activity of auranofin was examined in a Caenorhabditis elegans animal model of Cryptococcus neoformans infection. Auranofin significantly reduced the fungal load in infected C. elegans. Collectively, the present study provides valuable evidence that auranofin has significant promise to be repurposed as a novel antifungal agent and may offer a safe, effective, and quick supplement to current approaches for treating fungal infections.
Collapse
Affiliation(s)
- Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University West Lafayette, IN, USA
| | - Matthew Maland
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles Los Angeles, CA, USA
| | - Haroon Mohammad
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University West Lafayette, IN, USA
| | - Pete E Pascuzzi
- Purdue University Libraries, Purdue UniversityWest Lafayette, IN, USA; Department of Biochemistry, Purdue UniversityWest Lafayette, IN, USA
| | - Larisa Avramova
- Bindley Bioscience Center, Purdue University West Lafayette, IN, USA
| | - Carla M Koehler
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles Los Angeles, CA, USA
| | - Tony R Hazbun
- Bindley Bioscience Center, Purdue UniversityWest Lafayette, IN, USA; Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue UniversityWest Lafayette, IN, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue UniversityWest Lafayette, IN, USA; Purdue Institute for Inflammation, Immunology, and Infectious DiseasesWest Lafayette, IN, USA
| |
Collapse
|
45
|
Schmidt C, Karge B, Misgeld R, Prokop A, Franke R, Brönstrup M, Ott I. Gold(I) NHC Complexes: Antiproliferative Activity, Cellular Uptake, Inhibition of Mammalian and Bacterial Thioredoxin Reductases, and Gram-Positive Directed Antibacterial Effects. Chemistry 2017; 23:1869-1880. [DOI: 10.1002/chem.201604512] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Claudia Schmidt
- Institute of Medicinal and Pharmaceutical Chemistry; Technische Universität Braunschweig; Beethovenstrasse 55 38106 Braunschweig Germany
| | - Bianka Karge
- Department of Chemical Biology; Helmholtz Centre for Infection Research GmbH; Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Rainer Misgeld
- Department of Paedriatric Oncology; Children's Hospital Cologne; Amsterdamer Strasse 59 50735 Cologne Germany
| | - Aram Prokop
- Department of Paedriatric Oncology; Children's Hospital Cologne; Amsterdamer Strasse 59 50735 Cologne Germany
| | - Raimo Franke
- Department of Chemical Biology; Helmholtz Centre for Infection Research GmbH; Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Mark Brönstrup
- Department of Chemical Biology; Helmholtz Centre for Infection Research GmbH; Inhoffenstrasse 7 38124 Braunschweig Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry; Technische Universität Braunschweig; Beethovenstrasse 55 38106 Braunschweig Germany
| |
Collapse
|
46
|
In vitro antibacterial and time kill evaluation of mononuclear phosphanegold(I) dithiocarbamates. J Inorg Biochem 2016; 163:68-80. [DOI: 10.1016/j.jinorgbio.2016.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/26/2016] [Accepted: 08/01/2016] [Indexed: 11/22/2022]
|
47
|
Screening a Commercial Library of Pharmacologically Active Small Molecules against Staphylococcus aureus Biofilms. Antimicrob Agents Chemother 2016; 60:5663-72. [PMID: 27401577 DOI: 10.1128/aac.00377-16] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 07/06/2016] [Indexed: 12/25/2022] Open
Abstract
It is now well established that bacterial infections are often associated with biofilm phenotypes that demonstrate increased resistance to common antimicrobials. Further, due to the collective attrition of new antibiotic development programs by the pharmaceutical industries, drug repurposing is an attractive alternative. In this work, we screened 1,280 existing commercially available drugs in the Prestwick Chemical Library, some with previously unknown antimicrobial activity, against Staphylococcus aureus, one of the commonly encountered causative pathogens of burn and wound infections. From the primary screen of the entire Prestwick Chemical Library at a fixed concentration of 10 μM, 104 drugs were found to be effective against planktonic S. aureus strains, and not surprisingly, these were mostly antimicrobials and antiseptics. The activity of 18 selected repurposing candidates, that is, drugs that show antimicrobial activity that are not already considered antimicrobials, observed in the primary screen was confirmed in dose-response experiments. Finally, a subset of nine of these drug candidates was tested against preformed biofilms of S. aureus We found that three of these drugs, niclosamide, carmofur, and auranofin, possessed antimicrobial activity against preformed biofilms, making them attractive candidates for repurposing as novel antibiofilm therapies.
Collapse
|
48
|
Wiederhold NP, Patterson TF, Srinivasan A, Chaturvedi AK, Fothergill AW, Wormley FL, Ramasubramanian AK, Lopez-Ribot JL. Repurposing auranofin as an antifungal: In vitro activity against a variety of medically important fungi. Virulence 2016; 8:138-142. [PMID: 27268469 DOI: 10.1080/21505594.2016.1196301] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Repositioning old drugs can significantly decrease the time and effort that it takes to develop novel antifungal therapeutics, which represents a pressing and unmet clinical need due to the devastating nature of fungal infections. We have previously described the activity of auranofin, a gold thiol compound used to treat rheumatoid arthritis, against Candida albicans biofilms. Here we evaluate its antifungal spectrum of action and describe its activity against a variety of medically important fungi.
Collapse
Affiliation(s)
- Nathan P Wiederhold
- a Department of Pathology , The University of Texas Health Science Center at San Antonio , San Antonio , TX , USA.,b Department of Medicine , Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Thomas F Patterson
- b Department of Medicine , Division of Infectious Diseases, The University of Texas Health Science Center at San Antonio , San Antonio , TX , USA.,c South Texas Veterans Health Care System , San Antonio , TX , USA
| | - Anand Srinivasan
- d Department of Biomedical Engineering , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA.,e South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio , San Antonio , TX , USA
| | - Ashok K Chaturvedi
- e South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio , San Antonio , TX , USA.,f Department of Biology , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Annette W Fothergill
- a Department of Pathology , The University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Floyd L Wormley
- e South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio , San Antonio , TX , USA.,f Department of Biology , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Anand K Ramasubramanian
- d Department of Biomedical Engineering , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA.,e South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio , San Antonio , TX , USA
| | - José L Lopez-Ribot
- e South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio , San Antonio , TX , USA.,f Department of Biology , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| |
Collapse
|
49
|
Antibacterial activity and mechanism of action of auranofin against multi-drug resistant bacterial pathogens. Sci Rep 2016; 6:22571. [PMID: 26936660 PMCID: PMC4776257 DOI: 10.1038/srep22571] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/17/2016] [Indexed: 11/18/2022] Open
Abstract
Traditional methods employed to discover new antibiotics are both a time-consuming and financially-taxing venture. This has led researchers to mine existing libraries of clinical molecules in order to repurpose old drugs for new applications (as antimicrobials). Such an effort led to the discovery of auranofin, a drug initially approved as an anti-rheumatic agent, which also possesses potent antibacterial activity in a clinically achievable range. The present study demonstrates auranofin’s antibacterial activity is a complex process that involves inhibition of multiple biosynthetic pathways including cell wall, DNA, and bacterial protein synthesis. We also confirmed that the lack of activity of auranofin observed against Gram-negative bacteria is due to the permeability barrier conferred by the outer membrane. Auranofin’s ability to suppress bacterial protein synthesis leads to significant reduction in the production of key methicillin-resistant Staphylococcus aureus (MRSA) toxins. Additionally, auranofin is capable of eradicating intracellular MRSA present inside infected macrophage cells. Furthermore, auranofin is efficacious in a mouse model of MRSA systemic infection and significantly reduces the bacterial load in murine organs including the spleen and liver. Collectively, this study provides valuable evidence that auranofin has significant promise to be repurposed as a novel antibacterial for treatment of invasive bacterial infections.
Collapse
|
50
|
Abstract
BACKGROUND We identified auranofin as an antimicrobial compound utilizing a high-throughput screen using a Caenorhabditis elegans-Staphylococcus aureus infection model. Results/methodology: Treatment of infected nematodes with auranofin resulted in a prolonged survival rate of 95%, reached with 0.78 μg/ml. Further investigation of the antimicrobial activity of auranofin found inhibition against S. aureus, Enterococcus faecium and Enterococcus faecalis. Importantly, the fungal pathogens Cryptococcus neoformans was also effectively inhibited with an MIC at 0.5 μg/ml. Auranofin appears to target the thioredoxin system. CONCLUSION This work provides extensive additional data on the antibacterial effects of auranofin that includes both reference and clinical isolates and reports a novel inhibition of fungal pathogens by this compound.
Collapse
|