1
|
Quadros Barsé L, Ulfig A, Varatnitskaya M, Vázquez-Hernández M, Yoo J, Imann AM, Lupilov N, Fischer M, Becker K, Bandow JE, Leichert LI. Comparison of the mechanism of antimicrobial action of the gold(I) compound auranofin in Gram-positive and Gram-negative bacteria. Microbiol Spectr 2024; 12:e0013824. [PMID: 39377597 PMCID: PMC11537011 DOI: 10.1128/spectrum.00138-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 10/09/2024] Open
Abstract
While highly effective at killing Gram-positive bacteria, auranofin lacks significant activity against Gram-negative species for reasons that largely remain unclear. Here, we aimed to elucidate the molecular mechanisms underlying the low susceptibility of the Gram-negative model organism Escherichia coli to auranofin when compared to the Gram-positive model organism Bacillus subtilis. The proteome response of E. coli exposed to auranofin suggests a combination of inactivation of thiol-containing enzymes and the induction of systemic oxidative stress. Susceptibility tests in E. coli mutants lacking proteins upregulated upon auranofin treatment suggested that none of them are directly involved in E. coli's high tolerance to auranofin. E. coli cells lacking the efflux pump component TolC were more sensitive to auranofin treatment, but not to an extent that would fully explain the observed difference in susceptibility of Gram-positive and Gram-negative organisms. We thus tested whether E. coli's thioredoxin reductase (TrxB) is inherently less sensitive to auranofin than TrxB from B. subtilis, which was not the case. However, E. coli strains lacking the low-molecular-weight thiol glutathione, but not glutathione reductase, showed a high susceptibility to auranofin. Bacterial cells expressing the genetically encoded redox probe roGFP2 allowed us to observe the oxidation of cellular protein thiols in situ. Based on our findings, we hypothesize that auranofin leads to a global disturbance in the cellular thiol redox homeostasis in bacteria, but Gram-negative bacteria are inherently more resistant due to the presence of drug export systems and high cellular concentrations of glutathione.IMPORTANCEAuranofin is an FDA-approved drug for the treatment of rheumatoid arthritis. However, it has also high antibacterial activity, in particular against Gram-positive organisms. In the current antibiotics crisis, this would make it an ideal candidate for drug repurposing. However, its much lower activity against Gram-negative organisms prevents its broad-spectrum application. Here we show that, on the level of the presumed target, there is no difference in susceptibility between Gram-negative and Gram-positive species: thioredoxin reductases from both Escherichia coli and Bacillus subtilis are equally inhibited by auranofin. In both species, auranofin treatment leads to oxidative protein modification on a systemic level, as monitored by proteomics and the genetically encoded redox probe roGFP2. The single largest contributor to E. coli's relative resistance to auranofin seems to be the low-molecular-weight thiol glutathione, which is absent in B. subtilis and other Gram-positive species.
Collapse
Affiliation(s)
- Laísa Quadros Barsé
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Agnes Ulfig
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Marharyta Varatnitskaya
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | | | - Jihyun Yoo
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Astrid M. Imann
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
- Institute of Electrical Engineering and Applied Sciences–Molecular Biology, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - Natalie Lupilov
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| | - Marina Fischer
- Interdisciplinary Research Center, Justus Liebig University Giessen, Giessen, Germany
| | - Katja Becker
- Interdisciplinary Research Center, Justus Liebig University Giessen, Giessen, Germany
| | - Julia E. Bandow
- Faculty of Biology and Biotechnology, Applied Microbiology, Ruhr University Bochum, Bochum, Germany
| | - Lars I. Leichert
- Medical Faculty, Institute of Biochemistry and Pathobiochemistry–Microbial Biochemistry, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
2
|
Anjou C, Royer M, Bertrand É, Bredon M, Le Bris J, Salgueiro IA, Caulat LC, Dupuy B, Barbut F, Morvan C, Rolhion N, Martin-Verstraete I. Adaptation mechanisms of Clostridioides difficile to auranofin and its impact on human gut microbiota. NPJ Biofilms Microbiomes 2024; 10:86. [PMID: 39284817 PMCID: PMC11405772 DOI: 10.1038/s41522-024-00551-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Auranofin (AF), a former rheumatoid polyarthritis treatment, gained renewed interest for its use as an antimicrobial. AF is an inhibitor of thioredoxin reductase (TrxB), a thiol and protein repair enzyme, with an antibacterial activity against several bacteria including C. difficile, an enteropathogen causing post-antibiotic diarrhea. Several studies demonstrated the effect of AF on C. difficile physiology, but the crucial questions of resistance mechanisms and impact on microbiota remain unaddressed. We explored potential resistance mechanisms by studying the impact of TrxB multiplicity and by generating and characterizing adaptive mutations. We showed that if mutants inactivated for trxB genes have a lower MIC of AF, the number of TrxBs naturally present in clinical strains does not impact the MIC. All stable mutations isolated after AF long-term exposure were in the anti-sigma factor of σB and strongly affect physiology. Finally, we showed that AF has less impact on human gut microbiota than vancomycin.
Collapse
Affiliation(s)
- Cyril Anjou
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Marie Royer
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Unité Écologie et Évolution de la Résistance aux Antibiotiques, Paris, France
| | - Émilie Bertrand
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Marius Bredon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Julie Le Bris
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS UMR3525, Université Paris Cité, Paris, France
- Sorbonne Université, Collège Doctoral, École Doctorale Complexité du Vivant, 75005, Paris, France
| | - Iria Alonso Salgueiro
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Léo C Caulat
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Bruno Dupuy
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Frédéric Barbut
- Université Paris Cité, INSERM, UMR-1139, Paris, France
- National Reference Laboratory for C. difficile, Assistance Publique Hôpitaux de Paris, Hôpital Saint-Antoine, 75012, Paris, France
| | - Claire Morvan
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Nathalie Rolhion
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France.
- Institut Universitaire de France, Paris, France.
| |
Collapse
|
3
|
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
|
4
|
Ilyas M, Latif MS, Gul A, Babar MM, Rajadas J. Drug repurposing for bacterial infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:1-21. [PMID: 38942533 DOI: 10.1016/bs.pmbts.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Repurposing pharmaceuticals is a technique used to find new, alternate clinical applications for approved drug molecules. It may include altering the drug formulation, route of administration, dose or the dosage regimen. The process of repurposing medicines starts with screening libraries of previously approved drugs for the targeted disease condition. If after an the initial in silico, in vitro or in vivo experimentation, the molecule has been found to be active against a particular target, the molecule is considered as a good candidate for clinical trials. As the safety profile of such molecules is available from the previous data, significant time and resources are saved. These advantages of drug repurposing approach make it especially helpful for finding treatments for rapidly evolving conditions including bacterial infections. An ever-increasing incidence of antimicrobial resistance, owing to the mutations in bacterial genome, leads to therapeutic failure of many approved antibiotics. Repurposing the approved drug molecules for use as antibiotics can provide an effective means for the combating life-threatening bacterial diseases. A number of drugs have been considered for drug repurposing against bacterial infections. These include, but are not limited to, Auranofin, Closantel, and Toremifene that have been repurposed for various infections. In addition, the reallocation of route of administration, redefining dosage regimen and reformulation of dosage forms have also been carried out for repurposing purpose. The current chapter addresses the drug discovery and development process with relevance to repurposing against bacterial infections.
Collapse
Affiliation(s)
- Mahnoor Ilyas
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan; Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Muhammad Saad Latif
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Alvina Gul
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Mustafeez Mujtaba Babar
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan; Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, PaloAlto, CA, United States.
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute and Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford University, PaloAlto, CA, United States
| |
Collapse
|
5
|
Wu S, Chen Y, Chen Z, Wei F, Zhou Q, Li P, Gu Q. Reactive oxygen species and gastric carcinogenesis: The complex interaction between Helicobacter pylori and host. Helicobacter 2023; 28:e13024. [PMID: 37798959 DOI: 10.1111/hel.13024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023]
Abstract
Helicobacter pylori (H. pylori) is a highly successful human pathogen that colonizes stomach in around 50% of the global population. The colonization of bacterium induces an inflammatory response and a substantial rise in the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), mostly derived from host neutrophils and gastric epithelial cells, which play a crucial role in combating bacterial infections. However, H. pylori has developed various strategies to quench the deleterious effects of ROS, including the production of antioxidant enzymes, antioxidant proteins as well as blocking the generation of oxidants. The host's inability to eliminate H. pylori infection results in persistent ROS production. Notably, excessive ROS can disrupt the intracellular signal transduction and biological processes of the host, incurring chronic inflammation and cellular damage, such as DNA damage, lipid peroxidation, and protein oxidation. Markedly, the sustained inflammatory response and oxidative stress during H. pylori infection are major risk factor for gastric carcinogenesis. In this context, we summarize the literature on H. pylori infection-induced ROS production, the strategies used by H. pylori to counteract the host response, and subsequent host damage and gastric carcinogenesis.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Qingqing Zhou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
6
|
Johnstone MA, Holman MA, Self WT. Inhibition of selenoprotein synthesis is not the mechanism by which auranofin inhibits growth of Clostridioides difficile. Sci Rep 2023; 13:14733. [PMID: 37679389 PMCID: PMC10484987 DOI: 10.1038/s41598-023-36796-9] [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: 10/07/2022] [Accepted: 06/12/2023] [Indexed: 09/09/2023] Open
Abstract
Clostridioides difficile infections (CDIs) are responsible for a significant number of antibiotic-associated diarrheal cases. The standard-of-care antibiotics for C. difficile are limited to fidaxomicin and vancomycin, with the recently obsolete metronidazole recommended if both are unavailable. No new antimicrobials have been approved for CDI since fidaxomicin in 2011, despite varying rates of treatment failure among all standard-of-care drugs. Drug repurposing is a rational strategy to generate new antimicrobials out of existing therapeutics approved for other indications. Auranofin is a gold-containing anti-rheumatic drug with antimicrobial activity against C. difficile and other microbes. In a previous report, our group hypothesized that inhibition of selenoprotein biosynthesis was auranofin's primary mechanism of action against C. difficile. However, in this study, we discovered that C. difficile mutants lacking selenoproteins are still just as sensitive to auranofin as their respective wild-type strains. Moreover, we found that selenite supplementation dampens the activity of auranofin against C. difficile regardless of the presence of selenoproteins, suggesting that selenite's neutralization of auranofin is not because of compensation for a chemically induced selenium deficiency. Our results clarify the findings of our original study and may aid drug repurposing efforts in discovering the compound's true mechanism of action against C. difficile.
Collapse
Affiliation(s)
- Michael A Johnstone
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4110 Libra Drive, Orlando, FL, 32816, USA
| | - Matthew A Holman
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4110 Libra Drive, Orlando, FL, 32816, USA
| | - William T Self
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4110 Libra Drive, Orlando, FL, 32816, USA.
| |
Collapse
|
7
|
Ferrando R, Mitchell SG, Atrián-Blasco E, Cerrada E. Antibacterial properties of phosphine gold(I) complexes with 5-fluorouracil. Dalton Trans 2023. [PMID: 37448318 DOI: 10.1039/d3dt01159c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
New gold(I) complexes with coordination to 5-fluorouracil (5-FU), an anticancer drug with antibacterial properties, have been synthesised and characterised, and are the first reported examples of 5-FU-Au compounds. These new complexes show high solution stability, even in the presence of a cysteine derivative, and so were evaluated as antibacterial compounds against model Gram-positive and Gram-negative bacteria. All the complexes show excellent antibacterial activity against Gram-positive B. subtilis, most of them improving the activity of 5-FU alone. Furthermore, these new complexes are also active against Gram-negative E. coli, where [Au(5-FU)(PTA)], the complex with the smallest phosphane, is the most bactericidal, 32 times more active than 5-FU on its own.
Collapse
Affiliation(s)
- Ricardo Ferrando
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain.
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Scott G Mitchell
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena Atrián-Blasco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Gascón E, Otal I, Maisanaba S, Llana-Ruiz-Cabello M, Valero E, Repetto G, Jones PG, Oriol L, Jiménez J. Gold(I) metallocyclophosphazenes with antibacterial potency and antitumor efficacy. Synergistic antibacterial action of a heterometallic gold and silver-cyclophosphazene. Dalton Trans 2022; 51:13657-13674. [PMID: 36040292 DOI: 10.1039/d2dt01963a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
One of the most important uses of phosphazenes today involves its biomedical applications. They can also be employed as scaffolds for the design and construction of a variety of ligands in order to coordinate them to metallic drugs. The coordination chemistry of the (amino)cyclotriphosphazene ligand, [N3P3(NHCy)6], towards gold(I) complexes has been studied. Neutral complexes, [N3P3(NHCy)6{AuX}n] (X = Cl or C6F5; n = 1 or 2) (1-4), cationic complexes, [N3P3(NHCy)6{Au(PR3)}n](NO3)n (PR3 = PPh3, PPh2Me, TPA; n = 1, 2 or 3) (6-12) [TPA = 1,3,5-triaza-7-phosphaadamantane] and a heterometallic compound [N3P3(NHCy)6{Au(PPh3)}2{Ag(PPh3)}](NO3)3 (13) have been obtained and characterized by various methods including single-crystal X-ray diffraction for 7, which confirms the coordination of gold atoms to the nitrogens of the phosphazene ring. Compounds 1, 4, 6-13 were screened for in vitro cytotoxic activity against two tumor human cell lines, MCF7 (breast adenocarcinoma) and HepG2 (hepatocellular carcinoma), and for antimicrobial activity against five bacterial species including Gram-positive, Gram-negative, and Mycobacteria. Both the median inhibitory concentration (IC50) and minimum inhibitory concentration (MIC) values are among the lowest found for any gold or silver derivatives against the cell lines and particularly against the Gram-positive (S. aureus) strain and the mycobacteria used in this work. Structure-activity relationships are discussed in order to determine the influence of ancillary ligands and the number and type of metal atoms (silver or gold). Compounds 4 and 8 showed not only maximal potency on human cells but also some tumour selectivity. Remarkably, compound 13, with both gold and silver atoms, showed outstanding activity against both Gram-positive and Gram-negative strains (nanomolar range), thus having a cooperative effect between gold and silver, with MIC values which are similar or lower than those of gentamicine, ciprofloxacin and rifampicine. The broad spectrum antimicrobial efficacy of all these metallophosphazenes and particularly of heterometallic compound 13 could be very useful to obtain materials for surfaces with antimicrobial properties that are increasingly in demand.
Collapse
Affiliation(s)
- Elena Gascón
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain.
| | - Isabel Otal
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Universidad de Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Sara Maisanaba
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - María Llana-Ruiz-Cabello
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Eva Valero
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área Nutrición y Bromatología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Guillermo Repetto
- Departamento de Biología Molecular e Ingeniería Bioquímica, Área de Toxicología, Universidad Pablo de Olavide, Ctra. Utrera, Km 1, 41013 Sevilla, Spain
| | - Peter G Jones
- Institut für Anorganische und Analytische Chemie, Technische Universität Braunschweig, Hagenring 30, D-38106, Braunschweig, Germany
| | - Luis Oriol
- Departamento de Química Orgánica, Instituto de Nanociencia y Materiales de Aragón-Facultad de Ciencias, Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Josefina Jiménez
- Departamento de Química Inorgánica, Facultad de Ciencias, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Universidad de Zaragoza-C.S.I.C., Pedro Cerbuna 12, 50009 Zaragoza, Spain.
| |
Collapse
|
11
|
Tialiou A, Chin J, Keppler BK, Reithofer MR. Current Developments of N-Heterocyclic Carbene Au(I)/Au(III) Complexes toward Cancer Treatment. Biomedicines 2022; 10:biomedicines10061417. [PMID: 35740438 PMCID: PMC9219884 DOI: 10.3390/biomedicines10061417] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022] Open
Abstract
Since their first discovery, N-heterocyclic carbenes have had a significant impact on organometallic chemistry. Due to their nature as strong σ-donor and π-acceptor ligands, they are exceptionally well suited to stabilize Au(I) and Au(III) complexes in biological environments. Over the last decade, the development of rationally designed NHCAu(I/III) complexes to specifically target DNA has led to a new “gold rush” in bioinorganic chemistry. This review aims to summarize the latest advances of NHCAu(I/III) complexes that are able to interact with DNA. Furthermore, the latest advancements on acyclic diamino carbene gold complexes with anticancer activity are presented as these typically overlooked NHC alternatives offer great additional design possibilities in the toolbox of carbene-stabilized gold complexes for targeted therapy.
Collapse
Affiliation(s)
- Alexia Tialiou
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Jiamin Chin
- Institute of Inorganic Chemistry—Functional Materials, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
- Correspondence: (J.C.); (M.R.R.)
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Währinger Str. 42, 1090 Vienna, Austria
| | - Michael R. Reithofer
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Str. 42, 1090 Vienna, Austria; (A.T.); (B.K.K.)
- Correspondence: (J.C.); (M.R.R.)
| |
Collapse
|
12
|
Ndugire W, Raviranga NGH, Lao J, Ramström O, Yan M. Gold Nanoclusters as Nanoantibiotic Auranofin Analogues. Adv Healthc Mater 2022; 11:e2101032. [PMID: 34350709 PMCID: PMC8816973 DOI: 10.1002/adhm.202101032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/13/2021] [Indexed: 12/21/2022]
Abstract
Auranofin, a gold(I)-complex with tetraacetylated thioglucose (Ac4 GlcSH) and triethylphosphine ligands, is an FDA-approved drug used as an anti-inflammatory aid in the treatment of rheumatoid arthritis. In repurposing auranofin for other diseases, it was found that the drug showed significant activity against Gram-positive but was inactive against Gram-negative bacteria. Herein, the design and synthesis of gold nanoclusters (AuNCs) based on the structural motif of auranofin are reported. Phosphine-capped AuNCs are synthesized and glycosylated, yielding auranofin analogues with mixed triphenylphosphine monosulfonate (TPPMS)/Ac4 GlcSH ligand shells. These AuNCs are active against both Gram-negative and Gram-positive bacteria, including multidrug-resistant pathogens. Notably, an auranofin analogue, a mixed-ligand 1.6 nm AuNC 4b, is more active than auranofin against Pseudomonas aeruginosa, while exhibiting lower toxicity against human A549 cells. The enhanced antibacterial activity of these AuNCs is characterized by a greater uptake of Au by the bacteria compared to AuI complexes. Additional factors include increased oxidative stress, moderate inhibition of thioredoxin reductase (TrxR), and DNA damage. Most intriguingly, the uptake of AuNCs are not affected by the bacterial outer membrane (OM) barrier or by binding with the extracellular proteins. This contrasts with AuI complexes like auranofin that are susceptible to protein binding and hindered by the OM barrier.
Collapse
Affiliation(s)
- William Ndugire
- Department of Chemistry, University of Massachusetts Lowell, One University Ave., Lowell, MA, 01854, USA
| | - N G Hasitha Raviranga
- Department of Chemistry, University of Massachusetts Lowell, One University Ave., Lowell, MA, 01854, USA
| | - Jingzhe Lao
- Department of Chemistry, University of Massachusetts Lowell, One University Ave., Lowell, MA, 01854, USA
| | - Olof Ramström
- Department of Chemistry, University of Massachusetts Lowell, One University Ave., Lowell, MA, 01854, USA
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, SE-39182, Sweden
| | - Mingdi Yan
- Department of Chemistry, University of Massachusetts Lowell, One University Ave., Lowell, MA, 01854, USA
| |
Collapse
|
13
|
Jasni N, Saidin S, Arifin N, Azman DK, Shin LN, Othman N. A Review: Natural and Synthetic Compounds Targeting Entamoeba histolytica and Its Biological Membrane. MEMBRANES 2022; 12:membranes12040396. [PMID: 35448367 PMCID: PMC9024486 DOI: 10.3390/membranes12040396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/23/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
Amoebiasis is the third most common parasitic cause of morbidity and mortality, particularly in countries with poor hygienic settings in central and south America, Africa, and India. This disease is caused by a protozoan parasite, namely Entamoeba histolytica, which infects approximately 50 million people worldwide, resulting in 70,000 deaths every year. Since the 1960s, E. histolytica infection has been successfully treated with metronidazole. However, there are drawbacks to metronidazole therapy: the side effects, duration of treatment, and need for additional drugs to prevent transmission. Previous interdisciplinary studies, including biophysics, bioinformatics, chemistry, and, more recently, lipidomics studies, have increased biomembranes’ publicity. The biological membranes are comprised of a mixture of membrane and cytosolic proteins. They work hand in hand mainly at the membrane part. They act as dedicated platforms for a whole range of cellular processes, such as cell proliferation, adhesion, migration, and intracellular trafficking, thus are appealing targets for drug treatment. Therefore, this review aims to observe the updated trend of the research regarding the biological membranes of E. histolytica from 2015 to 2021, which may help further research regarding the drug targeting the biological membrane.
Collapse
Affiliation(s)
- Nurhana Jasni
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia; (N.J.); (N.A.); (D.K.A.); (L.N.S.)
| | - Syazwan Saidin
- Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, Tanjung Malim 35900, Malaysia;
| | - Norsyahida Arifin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia; (N.J.); (N.A.); (D.K.A.); (L.N.S.)
| | - Daruliza Kernain Azman
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia; (N.J.); (N.A.); (D.K.A.); (L.N.S.)
| | - Lai Ngit Shin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia; (N.J.); (N.A.); (D.K.A.); (L.N.S.)
| | - Nurulhasanah Othman
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia; (N.J.); (N.A.); (D.K.A.); (L.N.S.)
- Correspondence:
| |
Collapse
|
14
|
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]
|
15
|
New Auranofin Analogs with Antibacterial Properties against Burkholderia Clinical Isolates. Antibiotics (Basel) 2021; 10:antibiotics10121443. [PMID: 34943654 PMCID: PMC8697972 DOI: 10.3390/antibiotics10121443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/17/2022] Open
Abstract
Bacteria of the genus Burkholderia include pathogenic Burkholderia mallei, Burkholderia pseudomallei and the Burkholderia cepacia complex (Bcc). These Gram-negative pathogens have intrinsic drug resistance, which makes treatment of infections difficult. Bcc affects individuals with cystic fibrosis (CF) and the species B. cenocepacia is associated with one of the worst clinical outcomes. Following the repurposing of auranofin as an antibacterial against Gram-positive bacteria, we previously synthetized auranofin analogs with activity against Gram-negatives. In this work, we show that two auranofin analogs, MS-40S and MS-40, have antibiotic activity against Burkholderia clinical isolates. The compounds are bactericidal against B. cenocepacia and kill stationary-phase cells and persisters without selecting for multistep resistance. Caenorhabditis elegans and Galleria mellonella tolerated high concentrations of MS-40S and MS-40, demonstrating that these compounds have low toxicity in these model organisms. In summary, we show that MS-40 and MS-40S have antimicrobial properties that warrant further investigations to determine their therapeutic potential against Burkholderia infections.
Collapse
|
16
|
New Auranofin Analogs with Antibacterial Properties against Burkholderia Clinical Isolates. Antibiotics (Basel) 2021. [PMID: 34943654 DOI: 10.3390/antibiotics10121443/s1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Bacteria of the genus Burkholderia include pathogenic Burkholderia mallei, Burkholderia pseudomallei and the Burkholderia cepacia complex (Bcc). These Gram-negative pathogens have intrinsic drug resistance, which makes treatment of infections difficult. Bcc affects individuals with cystic fibrosis (CF) and the species B. cenocepacia is associated with one of the worst clinical outcomes. Following the repurposing of auranofin as an antibacterial against Gram-positive bacteria, we previously synthetized auranofin analogs with activity against Gram-negatives. In this work, we show that two auranofin analogs, MS-40S and MS-40, have antibiotic activity against Burkholderia clinical isolates. The compounds are bactericidal against B. cenocepacia and kill stationary-phase cells and persisters without selecting for multistep resistance. Caenorhabditis elegans and Galleria mellonella tolerated high concentrations of MS-40S and MS-40, demonstrating that these compounds have low toxicity in these model organisms. In summary, we show that MS-40 and MS-40S have antimicrobial properties that warrant further investigations to determine their therapeutic potential against Burkholderia infections.
Collapse
|
17
|
Auranofin: Past to Present, and repurposing. Int Immunopharmacol 2021; 101:108272. [PMID: 34731781 DOI: 10.1016/j.intimp.2021.108272] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 01/15/2023]
Abstract
Auranofin (AF), a gold compound, has been used to treat rheumatoid arthritis (RA) for more than 40 years; however, its mechanism of action remains unknown. We revealed that AF inhibited the induction of proinflammatory proteins and their mRNAs by the inflammatory stimulants, cyclooxygenase-2 and inducible nitric oxide synthase, and their upstream regulator, NF-κB. AF also activated the proteins peroxyredoxin-1, Kelch-like ECH-associated protein 1, and NF-E2-related factor 2, and inhibited thioredoxin reductase, all of which are involved in oxidative or electrophilic stress under physiological conditions. Although the cell membrane was previously considered to be permeable to AF because of its hydrophobicity, the mechanisms responsible for transporting AF into and out of cells as well as its effects on the uptake and excretion of other drugs have not yet been elucidated. Antibodies for cytokines have recently been employed in the treatment of RA, which has had an impact on the use of AF. Trials to repurpose AF as a risk-controlled agent to treat cancers or infectious diseases, including severe acute respiratory syndrome coronavirus 2/coronavirus disease 2019, are ongoing. Novel gold compounds are also under development as anti-cancer and anti-infection agents.
Collapse
|
18
|
Chakraborty P, Oosterhuis D, Bonsignore R, Casini A, Olinga P, Scheffers D. An Organogold Compound as Potential Antimicrobial Agent against Drug-Resistant Bacteria: Initial Mechanistic Insights. ChemMedChem 2021; 16:3060-3070. [PMID: 34181818 PMCID: PMC8518660 DOI: 10.1002/cmdc.202100342] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 01/07/2023]
Abstract
The rise of antimicrobial resistance has necessitated novel strategies to efficiently combat pathogenic bacteria. Metal-based compounds have been proven as a possible alternative to classical organic drugs. Here, we have assessed the antibacterial activity of seven gold complexes of different families. One compound, a cyclometalated Au(III) C^N complex, showed activity against Gram-positive bacteria, including multi-drug resistant clinical strains. The mechanism of action of this compound was studied in Bacillus subtilis. Overall, the studies point towards a complex mode of antibacterial action, which does not include induction of oxidative stress or cell membrane damage. A number of genes related to metal transport and homeostasis were upregulated upon short treatment of the cells with gold compound. Toxicity tests conducted on precision-cut mouse tissue slices ex vivo revealed that the organogold compound is poorly toxic to mouse liver and kidney tissues, and may thus, be treated as an antibacterial drug candidate.
Collapse
Affiliation(s)
- Parichita Chakraborty
- Department of Molecular MicrobiologyGroningen Institute for Biomolecular Sciences and BiotechnologyUniversity of Groningen9747 AGGroningenThe Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and BiopharmacyGroningen Research Institute of PharmacyUniversity of Groningen9713AVGroningenThe Netherlands
| | - Riccardo Bonsignore
- Chair of Medicinal and Bioinorganic ChemistryDepartment of ChemistryTechnical University of MunichLichtenbergstr. 485748Garching b. MünchenGermany
| | - Angela Casini
- Chair of Medicinal and Bioinorganic ChemistryDepartment of ChemistryTechnical University of MunichLichtenbergstr. 485748Garching b. MünchenGermany
| | - Peter Olinga
- Department of Pharmaceutical Technology and BiopharmacyGroningen Research Institute of PharmacyUniversity of Groningen9713AVGroningenThe Netherlands
| | - Dirk‐Jan Scheffers
- Department of Molecular MicrobiologyGroningen Institute for Biomolecular Sciences and BiotechnologyUniversity of Groningen9747 AGGroningenThe Netherlands
| |
Collapse
|
19
|
Rollin-Pinheiro R, Borba-Santos LP, da Silva Xisto MID, de Castro-Almeida Y, Rochetti VP, Rozental S, Barreto-Bergter E. Identification of Promising Antifungal Drugs against Scedosporium and Lomentospora Species after Screening of Pathogen Box Library. J Fungi (Basel) 2021; 7:jof7100803. [PMID: 34682224 PMCID: PMC8539698 DOI: 10.3390/jof7100803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/20/2022] Open
Abstract
Fungal infections have been increasing during the last decades. Scedosporium and Lomentospora species are filamentous fungi most associated to those infections, especially in immunocompromised patients. Considering the limited options of treatment and the emergence of resistant isolates, an increasing concern motivates the development of new therapeutic alternatives. In this context, the present study screened the Pathogen Box library to identify compounds with antifungal activity against Scedosporium and Lomentospora. Using antifungal susceptibility tests, biofilm analysis, scanning electron microscopy (SEM), and synergism assay, auranofin and iodoquinol were found to present promising repurposing applications. Both compounds were active against different Scedosporium and Lomentospora, including planktonic cells and biofilm. SEM revealed morphological alterations and synergism analysis showed that both drugs present positive interactions with voriconazole, fluconazole, and caspofungin. These data suggest that auranofin and iodoquinol are promising compounds to be studied as repurposing approaches against scedosporiosis and lomentosporiosis.
Collapse
Affiliation(s)
- Rodrigo Rollin-Pinheiro
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (M.I.D.d.S.X.); (Y.d.C.-A.); (V.P.R.)
| | - Luana Pereira Borba-Santos
- Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Mariana Ingrid Dutra da Silva Xisto
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (M.I.D.d.S.X.); (Y.d.C.-A.); (V.P.R.)
| | - Yuri de Castro-Almeida
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (M.I.D.d.S.X.); (Y.d.C.-A.); (V.P.R.)
| | - Victor Pereira Rochetti
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (M.I.D.d.S.X.); (Y.d.C.-A.); (V.P.R.)
| | - Sonia Rozental
- Programa de Biologia Celular e Parasitologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.P.B.-S.); (S.R.)
| | - Eliana Barreto-Bergter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.R.-P.); (M.I.D.d.S.X.); (Y.d.C.-A.); (V.P.R.)
- Correspondence: ; Tel.: +55-(21)-3938-6741
| |
Collapse
|
20
|
Lata S, Akif M. Structure-based identification of natural compound inhibitor against M. tuberculosis thioredoxin reductase: insight from molecular docking and dynamics simulation. J Biomol Struct Dyn 2021; 39:4480-4489. [PMID: 32567497 DOI: 10.1080/07391102.2020.1778530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/30/2020] [Indexed: 12/19/2022]
Abstract
Antioxidant systems of M. tuberculosis (Mtb) play an important role in providing resistance in the hostile environment of mononuclear phagocytes. Thioredoxin system is a known antioxidant system that consists of three copies of thioredoxins (Trxs) and a single copy of thioredoxin reductase (TrxR). TrxR has been validated as an essential gene known to be involved in the reduction of peroxides, dinitrobenzenes and hydroperoxides, and is crucial in maintaining the survival of Mtb in macrophages. Recently, it has been demonstrated to be a druggable target. In this study, molecular docking was applied to screen more than 20,000 natural compounds from the Traditional Chinese Medicine database. Theoretical calculation of ΔGbinding by the Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) methods indicated two top-hit compounds that bind with a high affinity to the allosteric site, consisting of a hinge region, of TrxR. Further, stability and binding analysis of both compounds were carried out with molecular dynamics simulation. An analysis of conformational variation by principal component analysis (PCA) and protein contact network (PCN) uncovered the conformational changes in the compound-bound forms of protein. The NADPH domain formed many new interactions with the FAD domain in the compound-bound form, signifying that the binding may render an effect on the protein structure and function. Our results suggest that these two compounds could potentially be used for structure-based lead inhibitors against TrxR. The inhibitor selected as lead compound will be used further as a scaffold to optimize as novel anti-tuberculosis therapeutic.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Surabhi Lata
- Laboratory of Structural Biology, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana, India
| | - Mohd Akif
- Laboratory of Structural Biology, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana, India
| |
Collapse
|
21
|
Abstract
Gold compounds have been employed throughout history to treat various types of disease, from ancient times to the present day. In the year 1985, auranofin, a gold-containing compound, was approved by U.S. Food and Drug Administration (FDA) as a therapeutic agent to target rheumatoid arthritis that would facilitate easy oral drug administration as opposed to conventional intramuscular injection used in treatments. Furthermore, auranofin demonstrates promising results for the treatment of various diseases beyond rheumatoid arthritis, including cancer, neurodegenerative diseases, acquired immune deficiency syndrome, and bacterial and parasitic infections. Various potential novel applications for auranofin have been proposed for treating human diseases. Auranofin has previously been demonstrated to inhibit thioredoxin reductase (TrxR) involved within the thioredoxin (Trx) system that comprises one of the critical cellular redox systems within the body. TrxR comprises the sole known enzyme that catalyzes Trx reduction. With cancers in particular, TrxR inhibition facilitates an increase in cellular oxidative stress and suppresses tumor growth. In this review, we describe the potential of auranofin to serve as an anticancer agent and further drug repurposing to utilize this as a strategy for further appropriate drug developments.
Collapse
Affiliation(s)
- Isao Momose
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| | - Takefumi Onodera
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation
| |
Collapse
|
22
|
Felix L, Mylonakis E, Fuchs BB. Thioredoxin Reductase Is a Valid Target for Antimicrobial Therapeutic Development Against Gram-Positive Bacteria. Front Microbiol 2021; 12:663481. [PMID: 33936021 PMCID: PMC8085250 DOI: 10.3389/fmicb.2021.663481] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
There is a drought of new antibacterial compounds that exploit novel targets. Thioredoxin reductase (TrxR) from the Gram-positive bacterial antioxidant thioredoxin system has emerged from multiple screening efforts as a potential target for auranofin, ebselen, shikonin, and allicin. Auranofin serves as the most encouraging proof of concept drug, demonstrating TrxR inhibition can result in bactericidal effects and inhibit Gram-positive bacteria in both planktonic and biofilm states. Minimal inhibitory concentrations are on par or lower than gold standard medications, even among drug resistant isolates. Importantly, existing drug resistance mechanisms that challenge treatment of infections like Staphylococcus aureus do not confer resistance to TrxR targeting compounds. The observed inhibition by multiple compounds and inability to generate a bacterial genetic mutant demonstrate TrxR appears to play an essential role in Gram-positive bacteria. These findings suggest TrxR can be exploited further for drug development. Examining the interaction between TrxR and these proof of concept compounds illustrates that compounds representing a new antimicrobial class can be developed to directly interact and inhibit the validated target.
Collapse
Affiliation(s)
- LewisOscar Felix
- 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
| |
Collapse
|
23
|
O'Loughlin J, Napolitano S, Alkhathami F, O'Beirne C, Marhöfer D, O'Shaughnessy M, Howe O, Tacke M, Rubini M. The Antibacterial Drug Candidate SBC3 is a Potent Inhibitor of Bacterial Thioredoxin Reductase. Chembiochem 2020; 22:1093-1098. [PMID: 33170522 DOI: 10.1002/cbic.202000707] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Indexed: 01/06/2023]
Abstract
Antibiotic resistance is a growing problem for public health and associated with increasing economic costs and mortality rates. Silver and silver-related compounds have been used for centuries due to their antimicrobial properties. In this work, we show that 1,3-dibenzyl-4,5-diphenyl-imidazol-2-ylidene silver(I) acetate/NHC*-Ag-OAc (SBC3) is a reversible, high affinity inhibitor of E. coli thioredoxin reductase (TrxR; Ki =10.8±1.2 nM). Minimal inhibition concentration (MIC) tests with different E. coli and P. aeruginosa strains demonstrated that SBC3 can efficiently inhibit bacterial cell growth, especially in combination with established antibiotics like gentamicin. Our results show that SBC3 is a promising antibiotic drug candidate targeting bacterial TrxR.
Collapse
Affiliation(s)
- Jennie O'Loughlin
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| | - Silvia Napolitano
- Department of Molecular Biology and Biophysics, ETH, Otto-Stern-Weg 5, 8093, Zürich, Switzerland
| | - Fahad Alkhathami
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| | - Cillian O'Beirne
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| | - Daniel Marhöfer
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| | - Megan O'Shaughnessy
- School of Biological and Health Sciences, Technical University Dublin, City Campus, Dublin, Ireland
| | - Orla Howe
- School of Biological and Health Sciences, Technical University Dublin, City Campus, Dublin, Ireland
| | - Matthias Tacke
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| | - Marina Rubini
- School of Chemistry, University College Dublin Belfield, Dublin 4, Ireland
| |
Collapse
|
24
|
Minori K, Rosa LB, Bonsignore R, Casini A, Miguel DC. Comparing the Antileishmanial Activity of Gold(I) and Gold(III) Compounds in L. amazonensis and L. braziliensis in Vitro. ChemMedChem 2020; 15:2146-2150. [PMID: 32830445 PMCID: PMC7756297 DOI: 10.1002/cmdc.202000536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 12/13/2022]
Abstract
A series of mononuclear coordination or organometallic AuI /AuIII complexes (1-9) have been comparatively studied in vitro for their antileishmanial activity against promastigotes and amastigotes, the clinically relevant parasite form, of Leishmania amazonensis and Leishmania braziliensis. One of the cationic AuI bis-N-heterocyclic carbenes (3) has low EC50 values (ca. 4 μM) in promastigotes cells and no toxicity in host macrophages. Together with two other AuIII complexes (6 and 7), the compound is also extremely effective in intracellular amastigotes from L. amazonensis. Initial mechanistic studies include an evaluation of the gold complexes' effect on L. amazonensis' plasma membrane integrity.
Collapse
Affiliation(s)
- Karen Minori
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| | - Letícia B. Rosa
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| | - Riccardo Bonsignore
- Department of ChemistryTechnical University of Munich (TUM)Lichtenbergstraße 485748Garching b. MünchenGermany
| | - Angela Casini
- Department of ChemistryTechnical University of Munich (TUM)Lichtenbergstraße 485748Garching b. MünchenGermany
| | - Danilo C. Miguel
- Department of Animal Biology, Biology InstituteUniversity of Campinas (UNICAMP)Rua Monteiro Lobato, 25513083-862.CampinasSPBrazil
| |
Collapse
|
25
|
Krzyżek P, Paluch E, Gościniak G. Synergistic Therapies as a Promising Option for the Treatment of Antibiotic-Resistant Helicobacter pylori. Antibiotics (Basel) 2020; 9:antibiotics9100658. [PMID: 33007899 PMCID: PMC7599531 DOI: 10.3390/antibiotics9100658] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Helicobacter pylori is a Gram-negative bacterium responsible for the development of gastric diseases. The issue of spreading antibiotic resistance of H. pylori and its limited therapeutic options is an important topic in modern gastroenterology. This phenomenon is greatly associated with a very narrow range of antibiotics used in standard therapies and, as a consequence, an alarmingly high detection of multidrug-resistant H. pylori strains. For this reason, scientists are increasingly focused on the search for new substances that will not only exhibit antibacterial effect against H. pylori, but also potentiate the activity of antibiotics. The aim of the current review is to present scientific reports showing newly discovered or repurposed compounds with an ability to enhance the antimicrobial activity of classically used antibiotics against H. pylori. To gain a broader context in their future application in therapies of H. pylori infections, their antimicrobial properties, such as minimal inhibitory concentrations and minimal bactericidal concentrations, dose- and time-dependent mode of action, and, if characterized, anti-biofilm and/or in vivo activity are further described. The authors of this review hope that this article will encourage the scientific community to expand research on the important issue of synergistic therapies in the context of combating H. pylori infections.
Collapse
|
26
|
Riches A, Hart CJS, Trenholme KR, Skinner-Adams TS. Anti- Giardia Drug Discovery: Current Status and Gut Feelings. J Med Chem 2020; 63:13330-13354. [PMID: 32869995 DOI: 10.1021/acs.jmedchem.0c00910] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Giardia parasites are ubiquitous protozoans of global importance that impact a wide range of animals including humans. They are the most common enteric pathogen of cats and dogs in developed countries and infect ∼1 billion people worldwide. While Giardia infections can be asymptomatic, they often result in severe and chronic diseases. There is also mounting evidence that they are linked to postinfection disorders. Despite growing evidence of the widespread morbidity associated with Giardia infections, current treatment options are limited to compound classes with broad antimicrobial activity. Frontline anti-Giardia drugs are also associated with increasing drug resistance and treatment failures. To improve the health and well-being of millions, new selective anti-Giardia drugs are needed alongside improved health education initiatives. Here we discuss current treatment options together with recent advances and gaps in drug discovery. We also propose criteria to guide the discovery of new anti-Giardia compounds.
Collapse
Affiliation(s)
- Andrew Riches
- Commonwealth Scientific and Industrial Research Organization, Biomedical Manufacturing, Clayton, Victoria 3168, Australia
| | - Christopher J S Hart
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia
| | - Katharine R Trenholme
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, Queensland 4029, Australia.,School of Medicine, University of Queensland, Brisbane, Queensland 4029, Australia
| | - Tina S Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland 4111, Australia
| |
Collapse
|
27
|
Abstract
Active efflux of antibiotics preventing their accumulation to toxic intracellular concentrations contributes to clinically relevant multidrug resistance. Inhibition of active efflux potentiates antibiotic activity, indicating that efflux inhibitors could be used in combination with antibiotics to reverse drug resistance. Expression of ramA by Salmonella enterica serovar Typhimurium increases in response to efflux inhibition, irrespective of the mode of inhibition. We hypothesized that measuring ramA promoter activity could act as a reporter of efflux inhibition. A rapid, inexpensive, and high-throughput green fluorescent protein (GFP) screen to identify efflux inhibitors was developed, validated, and implemented. Two chemical compound libraries were screened for compounds that increased GFP production. Fifty of the compounds in the 1,200-compound Prestwick chemical library were identified as potential efflux inhibitors, including the previously characterized efflux inhibitors mefloquine and thioridazine. There were 107 hits from a library of 47,168 proprietary compounds from L. Hoffmann La Roche; 45 were confirmed hits, and a dose response was determined. Dye efflux and accumulation assays showed that 40 Roche and three Prestwick chemical library compounds were efflux inhibitors. Most compounds had specific efflux-inhibitor-antibiotic combinations and/or species-specific synergy in antibiotic disc diffusion and checkerboard assays performed with Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, and Salmonella Typhimurium. These data indicate that both narrow-spectrum and broad-spectrum combinations of efflux inhibitors with antibiotics can be found. Eleven novel efflux inhibitor compounds potentiated antibiotic activities against at least one species of Gram-negative bacteria, and data revealing an E. coli mutant with loss of AcrB function suggested that these are AcrB inhibitors.IMPORTANCE Multidrug-resistant Gram-negative bacteria pose a serious threat to human and animal health. Molecules that inhibit multidrug efflux offer an alternative approach to resolving the challenges caused by antibiotic resistance, by potentiating the activity of old, licensed, and new antibiotics. We have developed, validated, and implemented a high-throughput screen and used it to identify efflux inhibitors from two compound libraries selected for their high chemical and pharmacological diversity. We found that the new high-throughput screen is a valuable tool to identify efflux inhibitors, as evidenced by the 43 new efflux inhibitors described in this study.
Collapse
|
28
|
Synthesis of Iron(II)-N-Heterocyclic Carbene Complexes: Paving the Way for a New Class of Antibiotics. Molecules 2020; 25:molecules25122917. [PMID: 32599931 PMCID: PMC7356145 DOI: 10.3390/molecules25122917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/09/2020] [Accepted: 06/22/2020] [Indexed: 11/17/2022] Open
Abstract
The synthesis and structural modulation of five pro-ligand salts was achieved using alternative sustainable synthetic strategies, the use of microwaves being the method of choice, with an 81% yield and an E factor of 43 for 3d. After complexation with Fe3(CO)12 by direct reaction with the appropriate pro-ligands at 130 °C, a set of iron(II) N-heterocyclic carbene (NHC) complexes were isolated and fully characterized (via 1H and 13C NMR and IR spectroscopy and elemental analysis). The antibacterial activities of the iron(II)-NHC complexes were tested against standard World Health Organization priority bacterial strains: Staphylococcus aureus ATCC 29213 and Escherichia coli ATCC 25922. The results showed a significant effect of the Fe(II)-NHC side-chain on the antibacterial activity against both Gram-negative and Gram-positive bacteria. Among all compounds, the most lipophilic iron complex, 3b, was found to be the most active one, with a minimum inhibitory concentration of 8 µg/mL. Pioneering mechanistic studies suggested an alternative mechanism of action (OH· formation), which opens the way for the development of a new class of antibiotics.
Collapse
|
29
|
Ouyang Y, Li J, Peng Y, Huang Z, Ren Q, Lu J. The Role and Mechanism of Thiol-Dependent Antioxidant System in Bacterial Drug Susceptibility and Resistance. Curr Med Chem 2020; 27:1940-1954. [DOI: 10.2174/0929867326666190524125232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 01/24/2019] [Accepted: 02/21/2019] [Indexed: 12/16/2022]
Abstract
Antibiotics play an irreplaceable role in the prevention and treatment of bacterial infection
diseases. However, because of the improper use of antibiotics, bacterial resistance emerges as a major
challenge of public health all over the world. The small thiol molecules such as glutathione can directly
react and conjugate with some antibiotics, which thus contribute to drug susceptibility and resistance.
Recently, accumulating evidence shows that there is a close link between the antibacterial activities of
some antibiotics and Reactive Oxygen Species (ROS). Thioredoxin and glutathione systems are two
main cellular disulfide reductase systems maintaining cellular ROS level. Therefore, these two thioldependent
antioxidant systems may affect the antibiotic susceptibility and resistance. Microorganisms
are equipped with different thiol-dependent antioxidant systems, which make the role of thioldependent
antioxidant systems in antibiotic susceptibility and resistance is different in various bacteria.
Here we will focus on the review on the advances of the effects of thiol-dependent antioxidant system
in the bacterial antibiotic susceptibility and resistance.
Collapse
Affiliation(s)
- Yanfang Ouyang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jing Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yi Peng
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhijun Huang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qiao Ren
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jun Lu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry, Ministry of Education (Southwest University), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
30
|
Antibiotics in the clinical pipeline in October 2019. J Antibiot (Tokyo) 2020; 73:329-364. [PMID: 32152527 PMCID: PMC7223789 DOI: 10.1038/s41429-020-0291-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/27/2022]
Abstract
The development of new and effective antibacterial drugs to treat multi-drug resistant (MDR) bacteria, especially Gram-negative (G−ve) pathogens, is acknowledged as one of the world’s most pressing health issues; however, the discovery and development of new, nontoxic antibacterials is not a straightforward scientific task, which is compounded by a challenging economic model. This review lists the antibacterials, β-lactamase/β-lactam inhibitor (BLI) combinations, and monoclonal antibodies (mAbs) first launched around the world since 2009 and details the seven new antibiotics and two new β-lactam/BLI combinations launched since 2016. The development status, mode of action, spectra of activity, lead source, and administration route for the 44 small molecule antibacterials, eight β-lactamase/BLI combinations, and one antibody drug conjugate (ADC) being evaluated in worldwide clinical trials at the end of October 2019 are described. Compounds discontinued from clinical development since 2016 and new antibacterial pharmacophores are also reviewed. There has been an increase in the number of early stage clinical candidates, which has been fueled by antibiotic-focused funding agencies; however, there is still a significant gap in the pipeline for the development of new antibacterials with activity against β-metallolactamases, orally administered with broad spectrum G−ve activity, and new treatments for MDR Acinetobacter and gonorrhea.
Collapse
|
31
|
Foerster S, Gustafsson TN, Brochado AR, Desilvestro V, Typas A, Unemo M. The first wide-scale drug repurposing screen using the Prestwick Chemical Library (1200 bioactive molecules) against Neisseria gonorrhoeae identifies high in vitro activity of auranofin and many additional drugs. APMIS 2020; 128:242-250. [PMID: 31811739 DOI: 10.1111/apm.13014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022]
Abstract
Treatment options for gonorrhoea are scarce. Drug repurposing of bioactive molecules approved for other conditions might therefore be of value. We developed a method for wide-scale, systematic drug repurposing screen to identify molecules with activity against Neisseria gonorrhoeae and screened the Prestwick Chemical Library (1200 FDA-approved drugs). As a proof-of-concept, we further examined one promising and interesting screening hit (auranofin; antirheumatic agent). Three WHO gonococcal reference strains (WHO F, O, P) were used for the Library screening. The strains were grown in presence of a fixed concentration of the library drugs in 384-well plates for 12 h, and the remaining bacterial respiration, to reflect growth, was then quantitatively measured using optical density (OD) 450 nm and a resazurin assay. The activity of auranofin was further examined using in vitro susceptibility testing (minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC)) against genetically diverse antimicrobial-resistant N. gonorrhoeae strains and time-kill assays. Sixty-eight molecules significantly inhibited bacterial growth of WHO F, O and P. Auranofin showed potent in vitro bactericidal activity (in MIC-, MBC- and time-kill assays) against four WHO reference strains. No cross-resistance between auranofin and any antimicrobial currently or previously used for gonorrhoea treatment was found when examining 51 selected antimicrobial-resistant gonococcal strains. In conclusion, this is the first wide-scale systematic screening effort for repurposing drugs for future treatment of gonorrhoea. Additional studies examining mechanism(s) of action, resistance development, in vivo anti-gonococcal activity and pharmacokinetics/pharmacodynamics for gonococcal infections of auranofin and several other significant screening hits would be valuable.
Collapse
Affiliation(s)
- Sunniva Foerster
- WHO Collaborating Centre for Gonorrhoea and other STIs, Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.,European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Tomas N Gustafsson
- Department of Clinical Microbiology, Sunderby Research Unit, Umeå University, Umeå, Sweden
| | - Anna Rita Brochado
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | | | - Athanasios Typas
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other STIs, Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
32
|
Dennis EK, Kim JH, Parkin S, Awuah SG, Garneau-Tsodikova S. Distorted Gold(I)–Phosphine Complexes as Antifungal Agents. J Med Chem 2019; 63:2455-2469. [DOI: 10.1021/acs.jmedchem.9b01436] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Emily K. Dennis
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| | - Jong Hyun Kim
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506-0055, United States
| | - Sean Parkin
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506-0055, United States
| | - Samuel G. Awuah
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, 505 Rose Street, Lexington, Kentucky 40506-0055, United States
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
33
|
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
|
34
|
Meier-Menches SM, Aikman B, Döllerer D, Klooster WT, Coles SJ, Santi N, Luk L, Casini A, Bonsignore R. Comparative biological evaluation and G-quadruplex interaction studies of two new families of organometallic gold(I) complexes featuring N-heterocyclic carbene and alkynyl ligands. J Inorg Biochem 2019; 202:110844. [PMID: 31739113 DOI: 10.1016/j.jinorgbio.2019.110844] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/07/2019] [Accepted: 09/08/2019] [Indexed: 12/24/2022]
Abstract
Experimental organometallic gold(I) compounds hold promise for anticancer therapy. This study reports the synthesis of two novel families of gold(I) complexes, including N1-substituted bis-N-heterocyclic carbene (NHC) complexes of general formula [Au(N1-TBM)2]BF4 (N1-TBM = N1-substituted 9-methyltheobromin-8-ylidene) and mixed gold(I) NHC-alkynyl complexes, [Au(N1-TBM)alkynyl]. The compounds were fully characterised for their structure and stability in aqueous environment and in the presence of N-acetyl cysteine by nuclear magnetic resonance (NMR) spectroscopy. The structures of bis(1-ethyl-3,7,9-trimethylxanthin-8-ylidene)gold(I), (4-ethynylpyridine)(1,9-dimethyltheobromine-8-ylidene)gold(I) and of (2,8-Diethyl-10-(4-ethynylphenyl)-5,5-difluoro-1,3,7,9-tetramethyl-5H-4λ4,5λ4-dipyrrolo[1,2-c:2',1'-f][1,3,2]diazaborinine)(1,3,7,9-tetramethylxanthin-8-ylidene)gold(I) were also confirmed by X-ray diffraction analysis. The compounds were studied for their properties as DNA G-quadruplex (G4 s) stabilizers by fluorescence resonance energy transfer (FRET) DNA melting. Only the cationic [Au(N1-TBM)2]BF4 family showed moderate G4 stabilization properties with respect to the previously reported benchmark compound [Au(9-methylcaffein-8-ylidene)2]+ (AuTMX2). However, the compounds also showed marked selectivity for binding to G4 structures with respect to duplex DNA in competition experiments. For selected complexes, the interactions with G4 s were also confirmed by circular dichroism (CD) studies. Furthermore, the gold(I) complexes were assessed for their antiproliferative effects in human cancer cells in vitro, displaying moderate activity. Of note, among the mixed gold(I) NHC-alkynyl compounds, one features a fluorescent boron-dipyrromethene (BODIPY) moiety which allowed determining its uptake into the cytoplasm of cancer cells by fluorescence microscopy.
Collapse
Affiliation(s)
- Samuel M Meier-Menches
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom
| | - Brech Aikman
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom
| | - Daniel Döllerer
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom
| | - Wim T Klooster
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Simon J Coles
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Nicolò Santi
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom
| | - Louis Luk
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom
| | - Angela Casini
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom; Department of Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany.
| | - Riccardo Bonsignore
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF103AT, United Kingdom.
| |
Collapse
|
35
|
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
|
36
|
Abhishek S, Sivadas S, Satish M, Deeksha W, Rajakumara E. Dynamic Basis for Auranofin Drug Recognition by Thiol-Reductases of Human Pathogens and Intermediate Coordinated Adduct Formation with Catalytic Cysteine Residues. ACS OMEGA 2019; 4:9593-9602. [PMID: 31460050 PMCID: PMC6649031 DOI: 10.1021/acsomega.9b00529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/14/2019] [Indexed: 05/13/2023]
Abstract
In all the living systems, reactive oxygen species (ROS) metabolism provides resistance against internal and external oxidative stresses. Auranofin (AF), an FDA-approved gold [Au(I)]-conjugated drug, is known to selectively target thiol-reductases, key enzymes involved in ROS metabolism. AF has been successfully tested for its inhibitory activity through biochemical studies, both in vitro and in vivo, against a diverse range of pathogens including protozoa, nematodes, bacteria, and so forth. Cocrystal structures of thiol-reductases complexed with AF revealed that Au(I) was coordinately linked to catalytic cysteines, but the mechanism of transfer of Au(I) from AF to catalytic cysteines still remains unknown. In this study, we have employed computational approaches to understand the interaction of AF with thiol-reductases of selected human pathogens. A similar network of interactions of AF was observed in all the studied enzymes. Also, we have shown that tailor-made analogues of AF can be designed against selective thiol-reductases for targeted inhibition. Molecular dynamics studies show that the AF-intermediates, tetraacetylthioglucose (TAG)-gold, and triethylphosphine (TP)-gold, coordinately linked to one of catalytic cysteines, remain stable in the binding pocket of thiol-reductases for Leishmania infantum and Plasmodium falciparum (PfTrxR). This suggests that the TP and TAG moieties of AF may be sequentially eliminated during the transfer of Au(I) to catalytic cysteines of the receptor.
Collapse
|
37
|
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
|
38
|
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
|
39
|
Elie BT, Fernández-Gallardo J, Curado N, Cornejo MA, Ramos JW, Contel M. Bimetallic titanocene-gold phosphane complexes inhibit invasion, metastasis, and angiogenesis-associated signaling molecules in renal cancer. Eur J Med Chem 2018; 161:310-322. [PMID: 30368130 DOI: 10.1016/j.ejmech.2018.10.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 01/08/2023]
Abstract
Following promising recent in vitro and in vivo studies of the anticancer efficacies of heterometallic titanocene-gold chemotherapeutic candidates against renal cancer, we report here on the synthesis, characterization, stability studies and biological evaluation of a new titanocene complex containing a gold-triethylphosphane fragment [(η-C5H5)2TiMe(μ-mba)Au(PEt3)] (4) Titanofin. The compound is more stable in physiological fluid than those previously reported, and it is highly cytotoxic against a line of human clear cell renal carcinoma. We describe here preliminary mechanistic data for this compound and previously reported [(η-C5H5)2TiMe(μ-mba)Au(PPh3)] (2) Titanocref which displayed remarkable activity in an in vivo mouse model. Mechanistic studies were carried out in the human clear cell renal carcinoma Caki-1 line for the bimetallic compounds [(η-C5H5)2TiMe(μ-mba)Au(PR3)] (PR3 = PPh32 Titanocref and PEt34 Titanofin), the two monometallic gold derivatives [Au(Hmba)(PR3)] (PR3 = PPh31 cref; PEt33 fin), titanocene dichloride and Auranofin as controls. These studies indicate that bimetallic compounds Titanocref (2) and Titanofin (4) are more cytotoxic than gold monometallic derivatives (1 and 3) and significantly more cytotoxic than titanocene dichloride while being quite selective. Titanocref (2) and Titanofin (4) inhibit migration, invasion, and angiogenic assembly along with molecular markers associated with these processes such as prometastatic IL(s), MMP(s), TNF-α, and proangiogenic VEGF, FGF-basic. The bimetallic compounds also strongly inhibit the mitochondrial protein TrxR often overexpressed in cancer cells evading apoptosis and also inhibit FOXC2, PECAM-1, and HIF-1α whose overexpression is linked to resistance to genotoxic chemotherapy. In summary, bimetallic titanocene-gold phosphane complexes (Titanocref 2 and Titanofin 4) are very promising candidates for further preclinical evaluations for the treatment of renal cancer.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA
| | - Jacob Fernández-Gallardo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Natalia Curado
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Mike A Cornejo
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, USA; Biology PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Chemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 Fifth Avenue, New York, NY, 10016, USA; Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, USA.
| |
Collapse
|
40
|
Medicinal Applications of Gold(I/III)-Based Complexes Bearing N-Heterocyclic Carbene and Phosphine Ligands. J Organomet Chem 2018. [DOI: 10.1016/j.jorganchem.2018.04.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Sweeney NL, Lipker L, Hanson AM, Bohl CJ, Engel KE, Kalous KS, Stemper ME, Sem DS, Schwan WR. Docking into Mycobacterium tuberculosis Thioredoxin Reductase Protein Yields Pyrazolone Lead Molecules for Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2017; 6:antibiotics6010004. [PMID: 28134858 PMCID: PMC5372984 DOI: 10.3390/antibiotics6010004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 01/20/2023] Open
Abstract
The thioredoxin/thioredoxin reductase system (Trx/TrxR) is an attractive drug target because of its involvement in a number of important physiological processes, from DNA synthesis to regulating signal transduction. This study describes the finding of pyrazolone compounds that are active against Staphylococcus aureus. Initially, the project was focused on discovering small molecules that may have antibacterial properties targeting the Mycobacterium tuberculosis thioredoxin reductase. This led to the discovery of a pyrazolone scaffold-containing compound series that showed bactericidal capability against S. aureus strains, including drug-resistant clinical isolates. The findings support continued development of the pyrazolone compounds as potential anti-S. aureus antibiotics.
Collapse
Affiliation(s)
- Noreena L Sweeney
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N Lake Shore Dr, Mequon, WI 53097, USA.
| | - Lauren Lipker
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, USA.
| | - Alicia M Hanson
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N Lake Shore Dr, Mequon, WI 53097, USA.
| | - Chris J Bohl
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N Lake Shore Dr, Mequon, WI 53097, USA.
| | - Katie E Engel
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, USA.
| | - Kelsey S Kalous
- Department of Biochemistry, Medical College of Wisconsin, Wauwatosa, WI 53226, USA.
| | - Mary E Stemper
- Marshfield Clinic Research Foundation, Marshfield, WI 54449, USA.
| | - Daniel S Sem
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N Lake Shore Dr, Mequon, WI 53097, USA.
| | - William R Schwan
- Department of Microbiology, University of Wisconsin-La Crosse, La Crosse, WI 54601, USA.
| |
Collapse
|