1
|
Rahmani F, Demirci G, Cortat Y, Crochet A, Zobi F. Antimicrobial Piano-Stool and Polypyridyl Ru(II) Complexes with Thiazolhidrazinylidene-Chroman-2,4-Dione: Tautomerism, Membrane Disruption, and Electron Transport Interference. Chembiochem 2025; 26:e202401025. [PMID: 40211911 DOI: 10.1002/cbic.202401025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/09/2025] [Indexed: 05/02/2025]
Abstract
A library of (η6-p-cymene)Ru(II) and Ru(II) bis-bpy complexes bearing thiazolhidrazinylidene-chroman-2,4-dione was synthesized, characterized, and the molecules are evaluated for their antibacterial activity and cytotoxicity. From this library, several compounds are identified as being active against Methicillin-resistant Staphylococcus aureus and Methicillin-sensitive S. aureus. A polypyridyl complex showed a noteworthy minimum inhibitor concentration of 3.1 μm and no toxicity in healthy eukaryotic cells with a therapeutic index of >32. Active complexes appear to exert their antibiotic activity by affecting both the permeabilization of the bacterial membrane and/or the electron transport chain.
Collapse
Affiliation(s)
- Fatlinda Rahmani
- Department of Chemistry, University of Fribourg, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Gozde Demirci
- Department of Chemistry, University of Fribourg, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Youri Cortat
- Department of Chemistry, University of Fribourg, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Aurélien Crochet
- Department of Chemistry, University of Fribourg, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Chemin Du Musée 9, 1700, Fribourg, Switzerland
| |
Collapse
|
2
|
Kushwaha R, Banerjee S. Design, development, and therapeutic applications of rhenium tricarbonyl complexes. Future Med Chem 2025:1-3. [PMID: 40371592 DOI: 10.1080/17568919.2025.2504324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Affiliation(s)
- Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, India
| |
Collapse
|
3
|
Bellapukonda SM, Bandela R, Singampalli A, Srikanth D, Kumar P, Nanduri S, Yaddanapudi VM. A systematic review on the anti-microbial activities and structure-activity relationship (SAR) of quinoxaline derivatives. Eur J Med Chem 2025; 289:117472. [PMID: 40048800 DOI: 10.1016/j.ejmech.2025.117472] [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: 11/19/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025]
Abstract
Anti-microbial resistance has become a serious global health issue affecting millions of people worldwide. Despite extensive drug discovery efforts aimed at identifying potent molecules for effective anti-microbial treatments, the emergence of superbugs remains a significant challenge. Thus, developing novel therapeutic agents is required to combat these evolving threats. The quinoxaline scaffold emerges as a promising heterocyclic framework for developing novel anti-microbial agents. It's simple, flexible structure, coupled with its bioisosteric relationship to extensively explored quinoline and naphthalene scaffolds, offers a potential avenue for circumventing bacterial resistance developed against these established classes. Hence it has sparked interest in researchers to develop novel antibiotics based on the quinoxaline core. This review focuses on the recent advances of quinoxaline derivatives as anti-microbial agents and their structure-activity relationship studies based on the literature published from 2015 to 2024. The systematic presentation of this information will assist researchers in identifying key substitution patterns around the quinoxaline nucleus, facilitating the development of structure-activity relationship (SAR), and guiding the design of novel anti-microbial drugs to combat the growing threat of anti-microbial resistance.
Collapse
Affiliation(s)
- Sri Mounika Bellapukonda
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Rani Bandela
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Danaboina Srikanth
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Pardeep Kumar
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India.
| |
Collapse
|
4
|
Pakeeraiah K, Swain PP, Sahoo A, Panda PK, Mahapatra M, Mal S, Sahoo RK, Sahu PK, Paidesetty SK. Multimodal antibacterial potency of newly designed and synthesized Schiff's/Mannich based coumarin derivatives: potential inhibitors of bacterial DNA gyrase and biofilm production. RSC Adv 2024; 14:31633-31647. [PMID: 39376521 PMCID: PMC11457008 DOI: 10.1039/d4ra05756b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024] Open
Abstract
The briskened urge to develop potential antibacterial candidates against multidrug-resistant pathogens has motivated the present research study. Herein, newly synthesized coumarin derivatives with azomethine and amino-methylated as the functional groups have been focused on their antibacterial efficacy. The study proposed two distinct series: 3-acetyl substituted coumarin derivatives, followed by the Schiff base approach (5a-5i), and formaldehyde-secondary cyclic amine-based derivatives (7a-7g), using the Mannich base approach, further the compounds have been confirmed through various spectral studies. Further, target-specific binding affinity has been affirmed via in silico study. In vitro antibacterial study suggested compounds 5d and 5f to be most effective against S. aureus and multidrug-resistant K. pneumoniae, with MIC values of 8 and 16 μg mL-1. Among them, the compounds 5d and 5f showed excellent binding scores against different bacterial gyrase compared to the standard novobiocin. Based on RMRS, RMSF, Rg, and H-bond plots, MD simulation study at 100 ns also suggested better stability of 5d inside gyraseB of E. coli than the complex of E. coli-GyrB-novobiocin. The toxicity and pharmacokinetic profiles showed favorable drug-likeness. Overall, systematic in vitro and in silico assessment suggested that multimodal antibacterial derivatives 5d and 5f strongly inhibit both bacterial DNA gyrase and biofilm formation of drug-resistant pathogens, suggesting their potency in mainstream antibacterial therapy.
Collapse
Affiliation(s)
- Kakarla Pakeeraiah
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Pragyan Paramita Swain
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Alaka Sahoo
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
- Department of Skin & VD, Institute of Medical Sciences, SUM Hospital, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
- Research and Development Division, Salixiras Research Private Limited Bhubaneswar Odisha India
| | - Preetesh Kumar Panda
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Monalisa Mahapatra
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Suvadeep Mal
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Rajesh Kumar Sahoo
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Pratap Kumar Sahu
- Department of Pharmacology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| | - Sudhir Kumar Paidesetty
- Medicinal Chemistry Research Laboratory, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University Bhubaneswar 751003 Odisha India
| |
Collapse
|
5
|
Fulgencio S, Scaccaglia M, Frei A. Exploration of Rhenium Bisquinoline Tricarbonyl Complexes for their Antibacterial Properties. Chembiochem 2024; 25:e202400435. [PMID: 38785033 DOI: 10.1002/cbic.202400435] [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: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Metal complexes have emerged as a promising source for novel classes of antibacterial agents to combat the rise of antimicrobial resistance around the world. In the exploration of the transition metal chemical space for novel metalloantibiotics, the rhenium tricarbonyl moiety has been identified as a promising scaffold. Here we have prepared eight novel rhenium bisquinoline tricarbonyl complexes and explored their antibacterial properties. Significant activity against both Gram-positive and Gram-negative bacteria was observed. However, all complexes also showed significant toxicity against human cells, putting into question the prospects of this specific rhenium compound class as metalloantibiotics. To better understand their biological effects, we conduct the first mode of action studies on rhenium bisquinoline complexes and show that they are able to form pores through bacterial membranes. Their straight-forward synthesis and tuneability suggests that further optimisation of this compound class could lead to compounds with enhanced bacterial specificity.
Collapse
Affiliation(s)
- Sofia Fulgencio
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
| | - Mirco Scaccaglia
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Angelo Frei
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
- Department of Chemistry, University of York, York, YO10 5DD, U.K
| |
Collapse
|
6
|
Romão CC, Mendes SS, Rebelo C, Carvalho SM, Saraiva LM. Antimicrobial and anticancer properties of carbon monoxide releasing molecules of the fac-[Re(CO) 3(N-N)L] + family. Dalton Trans 2024; 53:11009-11020. [PMID: 38874948 DOI: 10.1039/d4dt00978a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The toxicity profile of fac-[Re(CO)3(N-N)L]+ complexes against microbial and tumoral cells has been extensively studied, primarily focusing on modifications to the bidentate diimine (N-N) ligand. However, less attention has been paid to modifications of the axial ligand L, which is perpendicular to the Re-N-N plane. This study reveals that the high toxicity of the fac-[Re(CO)3(bpy)(Ctz)]+ complex may be attributed to the structural effect of the trityl (CPh3) group present in clotrimazole, as removal of phenyl rings causes a significant decrease in the activity against Staphylococcus aureus (S. aureus). Moreover, substitution of the 1-tritylimidazole ligand by the structurally related ligands PPh3 and PCy3 maintains similarly high activity levels. These findings contribute to understanding the interactions of toxic complexes with bacterial membranes, suggesting that the ligand structures play a crucial role in inhibiting cell wall synthesis processes, potentially including Lipid II synthesis. Compounds with Ph3E (E = C-imidazole; P) groups also showed to be 10 times more toxic than cisplatin against three mammalian cell lines (IC50: 2-4 μM). In contrast, the analogue 1-benzylimidazole and 1-tert-butylimidazole derivatives were as toxic as cisplatin. We observed that the decomposition of the [Re(I)(CO)3] fragment inside mammalian cell lines liberates CO, which is expected to exert biological effects. Therefore, compounds of this family possessing the structural motif Ph3E seem to combine high antimicrobial and antitumoral activities, the latter being much higher than that of cisplatin.
Collapse
Affiliation(s)
- Carlos C Romão
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Sofia S Mendes
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Cátia Rebelo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Sandra M Carvalho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| | - Lígia M Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal.
| |
Collapse
|
7
|
Scaccaglia M, Birbaumer MP, Pinelli S, Pelosi G, Frei A. Discovery of antibacterial manganese(i) tricarbonyl complexes through combinatorial chemistry. Chem Sci 2024; 15:3907-3919. [PMID: 38487233 PMCID: PMC10935722 DOI: 10.1039/d3sc05326a] [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: 10/09/2023] [Accepted: 01/23/2024] [Indexed: 03/17/2024] Open
Abstract
The continuous rise of antimicrobial resistance is a serious threat to human health and already causing hundreds of thousands of deaths each year. While natural products and synthetic organic small molecules have provided the majority of our current antibiotic arsenal, they are falling short in providing new drugs with novel modes of action able to treat multidrug resistant bacteria. Metal complexes have recently shown promising results as antimicrobial agents, but the number of studied compounds is still vanishingly small, making it difficult to identify promising compound classes or elucidate structure-activity relationships. To accelerate the pace of discovery we have applied a combinatorial chemistry approach to the synthesis of metalloantibiotics. Utilizing robust Schiff-base chemistry and combining 7 picolinaldehydes with 10 aniline derivatives, and 6 axial ligands, either imidazole/pyridine-based or solvent, we have prepared a library of 420 novel manganese tricarbonyl complexes. All compounds were evaluated for their antibacterial properties and 10 lead compounds were identified, re-synthesised and fully characterised. All 10 compounds showed high and broad activity against Gram-positive bacteria. The best manganese complex displayed low toxicity against human cells with a therapeutic index of >100. In initial mode of action studies, we show that it targets the bacterial membrane without inducing pore formation or depolarisation. Instead, it releases its carbon monoxide ligands around the membrane and inhibits the bacterial respiratory chain. This work demonstrates that large numbers of metal complexes can be accessed through combinatorial synthesis and evaluated for their antibacterial potential, allowing for the rapid identification of promising metalloantibiotic lead compounds.
Collapse
Affiliation(s)
- Mirco Scaccaglia
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma 43124 Parma Italy
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| | - Michael P Birbaumer
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| | - Silvana Pinelli
- Department of Medicine and Surgery, University of Parma Via Gramsci 14 43126 Parma Italy
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma 43124 Parma Italy
| | - Angelo Frei
- Department of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern Freiestrasse 3 3012 Bern Switzerland
| |
Collapse
|
8
|
Wang LQ, Zhang CY, Chen JJ, Lin WJ, Yu GY, Deng LS, Ji XR, Duan XM, Xiong YS, Jiang GJ, Wang JT, Liao XW, Liu LH. Ru-Based Organometallic Agents Bearing Phenyl Hydroxide: Synthesis and Antibacterial Mechanism Study against Staphylococcus aureus. ChemMedChem 2023; 18:e202300306. [PMID: 37527976 DOI: 10.1002/cmdc.202300306] [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: 06/13/2023] [Revised: 07/17/2023] [Accepted: 08/01/2023] [Indexed: 08/03/2023]
Abstract
The development of antimicrobial agents with novel model of actions is a promising strategy to combat multiple resistant bacteria. Here, three ruthenium-based complexes, which acted as potential antimicrobial agents, were synthesized and characterized. Importantly, three complexes all showed strong bactericidal potency against Staphylococcus aureus. In particular, the most active one has a MIC of 6.25 μg/mL. Mechanistic studies indicated that ruthenium complex killed S. aureus by releasing ROS and damaging the integrity of bacterial cell membrane. In addition, the most active complex not only could inhibit the biofilm formation and hemolytic toxin secretion of S. aureus, but also serve as a potential antimicrobial adjuvant as well, which showed synergistic effects with eight traditional antibiotics. Finally, both G. mellonella larva infection model and mouse skin infection model all demonstrated that ruthenium complex also showed significant efficacy against S. aureus in vivo. In summary, our study suggested that ruthenium-based complexes bearing a phenyl hydroxide are promising antimicrobial agents for combating S. aureus.
Collapse
Affiliation(s)
- L Q Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - C Y Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - J J Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - W J Lin
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - G Y Yu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - L S Deng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X R Ji
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X M Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Y S Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - G J Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - J T Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - X W Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - L H Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, 418000, China
| |
Collapse
|
9
|
Waters JE, Stevens-Cullinane L, Siebenmann L, Hess J. Recent advances in the development of metal complexes as antibacterial agents with metal-specific modes of action. Curr Opin Microbiol 2023; 75:102347. [PMID: 37467616 DOI: 10.1016/j.mib.2023.102347] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 07/21/2023]
Abstract
The mounting burden of antimicrobial resistance (AMR) is one of the most concerning threats to public health worldwide. With low economic incentives and a dwindling supply of new drugs in clinical pipelines, more innovative approaches to novel drug design and development are desperately required. Metal-based compounds are rapidly emerging as an alternative to organic drugs, as they have the ability to kill pathogens via metal-specific modes of action. We herein review recent advances in metal-based antibacterial agents, including metal complexes, metal ions and catalytic metallodrugs. The review concludes with a perspective on the rational design of metal-based antibiotics, and how we can exploit their unique properties to tackle AMR.
Collapse
Affiliation(s)
- Jessica E Waters
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lars Stevens-Cullinane
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lukas Siebenmann
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Jeannine Hess
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom.
| |
Collapse
|
10
|
Li W, Huang Y, Liu Y, Wang Z, Li S, Chen Y, Ye Y, Yin S, Lei J. Antibacterial performance of heterocyclic organobismuth (III) complexes based on bidentate C,O‐coordinating ligands: Synergism of ligand identity and coordination number. Appl Organomet Chem 2023; 37. [DOI: 10.1002/aoc.7141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/11/2023] [Indexed: 01/04/2025]
Abstract
A series of heterocyclic organobismuth (III) complexes based on bidentate C,O‐coordinating ligands were designed and synthesized as antimicrobials. Antibacterial assays showed that complexes of this type are more effective for Gram‐positive bacteria (Staphylococcus aureus, Staphylococcus epidermidis, and Enterococcus faecalis) than Gram‐negative ones (Escherichia coli and Pseudomonas aeruginosa). Their activities are especially relevant to the synergism of lipophilicity, geometry, and stability, which depends on both the identity of coordinating ligands and the coordination number at the bismuth center. By comparison, the hypervalent 14‐Bi‐6 species diarylbismuth nitrate (8) was found to exhibit the most potent inhibitory effect, together with a high degree of selectivity, which gives an IC50(LO2)/MIC(Staphylococcus aureus) ratio of up to 23.08. Time–kill analysis demonstrated that complex 8 is bacteriostatic at low concentrations while displaying significant bactericidal activity at high doses. The results of drug resistance experiments suggested that complex 8 can inhibit the formation of bacterial biofilm and consequently delay or prevent the development of drug resistance. Furthermore, complex 8 also showed high inhibition efficiency against several drug‐resistant Staphylococcus aureus, and the MIC values are within the range of 0.39–1.56 μM, thus indicating the lack of cross‐resistance between this organometallic compound and commonly used antibiotics.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| | - Yan Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| | - Yongping Liu
- School of Medicine Hunan University of Chinese Medicine Changsha China
| | - Zixiu Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| | - Shan Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| | - Yi Chen
- School of Medicine Hunan University of Chinese Medicine Changsha China
| | - Yifei Ye
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| | - Shuang‐Feng Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Advanced Catalytic Engineering Research Center of the Ministry of Education, College of Chemistry and Chemical Engineering Hunan University Changsha China
| | - Jian Lei
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease of the Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, College of Pharmacy Gannan Medical University Ganzhou China
| |
Collapse
|
11
|
S Mendes S, Sorg M, Luís CM, Fontinha D, Francisco D, Moita D, C Romão C, G Pinho M, Pimentel C, Prudêncio M, M Saraiva L. Conjugated carbon monoxide-releasing molecules have broad-spectrum antimicrobial activity. Future Med Chem 2023; 15:1037-1048. [PMID: 37458074 DOI: 10.4155/fmc-2023-0103] [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] [Indexed: 07/18/2023] Open
Abstract
Aim: To test the antimicrobial effect of carbon monoxide-releasing molecules (CORMs) conjugated with azoles on different microorganisms. Methods & results: We used broth microdilution, checkerboard and cytotoxicity assays, as well as imaging, fluorescence and bioluminescence experiments to study [Re(CO)3(2,2'-bipyridyl)(Ctz)]+ (also known as ReBpyCtz). ReBpyCtz exhibits a low minimum inhibitory concentration value, increases the intracellular formation of reactive oxygen species and causes significant alterations on Staphylococcus aureus's membrane. ReBpyCtz is active against fungi, having a more prolonged fungicidal effect on Candida glabrata than clotrimazole and is selectively active on blood-stage malaria parasites, at a concentration that is not toxic to kidney epithelial cells. Conclusion: Conjugated CORMs have the potential to be active against different types of pathogens, thus constituting a promising class of broad-spectrum antimicrobials.
Collapse
Affiliation(s)
- Sofia S Mendes
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Moritz Sorg
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Cláudia Malta Luís
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Denise Francisco
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Diana Moita
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Carlos C Romão
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Mariana G Pinho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Catarina Pimentel
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028, Lisboa, Portugal
| | - Lígia M Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157, Oeiras, Portugal
| |
Collapse
|
12
|
Cortat Y, Nedyalkova M, Schindler K, Kadakia P, Demirci G, Nasiri Sovari S, Crochet A, Salentinig S, Lattuada M, Steiner OM, Zobi F. Computer-Aided Drug Design and Synthesis of Rhenium Clotrimazole Antimicrobial Agents. Antibiotics (Basel) 2023; 12:antibiotics12030619. [PMID: 36978486 PMCID: PMC10044843 DOI: 10.3390/antibiotics12030619] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
In the context of the global health issue caused by the growing occurrence of antimicrobial resistance (AMR), the need for novel antimicrobial agents is becoming alarming. Inorganic and organometallic complexes represent a relatively untapped source of antibiotics. Here, we report a computer-aided drug design (CADD) based on a 'scaffold-hopping' approach for the synthesis and antibacterial evaluation of fac-Re(I) tricarbonyl complexes bearing clotrimazole (ctz) as a monodentate ligand. The prepared molecules were selected following a pre-screening in silico analysis according to modification of the 2,2'-bipyridine (bpy) ligand in the coordination sphere of the complexes. CADD pointed to chiral 4,5-pinene and 5,6-pinene bipyridine derivatives as the most promising candidates. The corresponding complexes were synthesized, tested toward methicillin-sensitive and -resistant S. aureus strains, and the obtained results evaluated with regard to their binding affinity with a homology model of the S. aureus MurG enzyme. Overall, the title species revealed very similar minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values as those of the reference compound used as the scaffold in our approach. The obtained docking scores advocate the viability of 'scaffold-hopping' for de novo design, a potential strategy for more cost- and time-efficient discovery of new antibiotics.
Collapse
Affiliation(s)
- Youri Cortat
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Miroslava Nedyalkova
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Kevin Schindler
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Parth Kadakia
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Gozde Demirci
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Sara Nasiri Sovari
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Aurelien Crochet
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Stefan Salentinig
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Marco Lattuada
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Olimpia Mamula Steiner
- Haute école d'Ingénierie et d'Architecture, University of Applied Sciences Western Switzerland HES-SO, Pérolles 80, 1700 Fribourg, Switzerland
| | - Fabio Zobi
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| |
Collapse
|
13
|
Nasiri Sovari S, Kolly I, Schindler K, Djuric A, Srdic-Rajic T, Crochet A, Pavic A, Zobi F. Synthesis, characterization, and in vivo evaluation of the anticancer activity of a series of 5- and 6-(halomethyl)-2,2'-bipyridine rhenium tricarbonyl complexes. Dalton Trans 2023; 52:6934-6944. [PMID: 36916301 DOI: 10.1039/d2dt04041g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
We report the synthesis, characterization, and in vivo evaluation of the anticancer activity of a series of 5- and 6-(halomethyl)-2,2'-bipyridine rhenium tricarbonyl complexes. The study was promoted in order to understand if the presence and position of a reactive halomethyl substituent on the diimine ligand system of fac-[Re(CO)3]+ species may be a key molecular feature for the design of active and non-toxic anticancer agents. Only compounds potentially able to undergo ligand-based alkylating reactions show significant antiproliferative activity against colorectal and pancreatic cell lines. Of the new species presented in this study, one compound (5-(chloromethyl)-2,2'-bipyridine derivative) shows significant inhibition of pancreatic tumour growth in vivo in zebrafish-Panc-1 xenografts. The complex is noticeably effective at 8 μM concentration, lower than its in vitro IC50 values, being also capable of inhibiting in vivo cancer cells dissemination.
Collapse
Affiliation(s)
- Sara Nasiri Sovari
- Department of Chemistry, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| | - Isabelle Kolly
- Department of Chemistry, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| | - Kevin Schindler
- Department of Chemistry, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| | - Ana Djuric
- Department of experimental oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, Beograd, Republic of Serbia
| | - Tatjana Srdic-Rajic
- Department of experimental oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, Beograd, Republic of Serbia
| | - Aurelien Crochet
- Department of Chemistry, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Republic of Serbia.
| | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland.
| |
Collapse
|
14
|
Abstract
Bacteria, similar to most organisms, have a love-hate relationship with metals: a specific metal may be essential for survival yet toxic in certain forms and concentrations. Metal ions have a long history of antimicrobial activity and have received increasing attention in recent years owing to the rise of antimicrobial resistance. The search for antibacterial agents now encompasses metal ions, nanoparticles and metal complexes with antimicrobial activity ('metalloantibiotics'). Although yet to be advanced to the clinic, metalloantibiotics are a vast and underexplored group of compounds that could lead to a much-needed new class of antibiotics. This Review summarizes recent developments in this growing field, focusing on advances in the development of metalloantibiotics, in particular, those for which the mechanism of action has been investigated. We also provide an overview of alternative uses of metal complexes to combat bacterial infections, including antimicrobial photodynamic therapy and radionuclide diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Angelo Frei
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| | - Anthony D Verderosa
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alysha G Elliott
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Johannes Zuegg
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Mark A T Blaskovich
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
15
|
Frei A, Elliott AG, Kan A, Dinh H, Bräse S, Bruce AE, Bruce MR, Chen F, Humaidy D, Jung N, King AP, Lye PG, Maliszewska HK, Mansour AM, Matiadis D, Muñoz MP, Pai TY, Pokhrel S, Sadler PJ, Sagnou M, Taylor M, Wilson JJ, Woods D, Zuegg J, Meyer W, Cain AK, Cooper MA, Blaskovich MAT. Metal Complexes as Antifungals? From a Crowd-Sourced Compound Library to the First In Vivo Experiments. JACS AU 2022; 2:2277-2294. [PMID: 36311838 PMCID: PMC9597602 DOI: 10.1021/jacsau.2c00308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
There are currently fewer than 10 antifungal drugs in clinical development, but new fungal strains that are resistant to most current antifungals are spreading rapidly across the world. To prevent a second resistance crisis, new classes of antifungal drugs are urgently needed. Metal complexes have proven to be promising candidates for novel antibiotics, but so far, few compounds have been explored for their potential application as antifungal agents. In this work, we report the evaluation of 1039 metal-containing compounds that were screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD). We show that 20.9% of all metal compounds tested have antimicrobial activity against two representative Candida and Cryptococcus strains compared with only 1.1% of the >300,000 purely organic molecules tested through CO-ADD. We identified 90 metal compounds (8.7%) that show antifungal activity while not displaying any cytotoxicity against mammalian cell lines or hemolytic properties at similar concentrations. The structures of 21 metal complexes that display high antifungal activity (MIC ≤1.25 μM) are discussed and evaluated further against a broad panel of yeasts. Most of these have not been previously tested for antifungal activity. Eleven of these metal complexes were tested for toxicity in the Galleria mellonella moth larva model, revealing that only one compound showed signs of toxicity at the highest injected concentration. Lastly, we demonstrated that the organo-Pt(II) cyclooctadiene complex Pt1 significantly reduces fungal load in an in vivo G. mellonella infection model. These findings showcase that the structural and chemical diversity of metal-based compounds can be an invaluable tool in the development of new drugs against infectious diseases.
Collapse
Affiliation(s)
- Angelo Frei
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
- Department
of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012Bern, Switzerland
| | - Alysha G. Elliott
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Alex Kan
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Hue Dinh
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Stefan Bräse
- Institute
of Organic Chemistry, Karlsruhe Institute
of Technology, Fritz-Haber-Weg 6, 76131Karlsruhe, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - Alice E. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Mitchell R. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Feng Chen
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Dhirgam Humaidy
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Nicole Jung
- Karlsruhe
Nano Micro Facility (KNMF), Karlsruhe Institute
of Technology, Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - A. Paden King
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Peter G. Lye
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Hanna K. Maliszewska
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Ahmed M. Mansour
- Chemistry
Department, Faculty of Science, Cairo University, Giza12613, Egypt
| | - Dimitris Matiadis
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - María Paz Muñoz
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Tsung-Yu Pai
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Shyam Pokhrel
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Marina Sagnou
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - Michelle Taylor
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Justin J. Wilson
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Dean Woods
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Johannes Zuegg
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Wieland Meyer
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Amy K. Cain
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Matthew A. Cooper
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| |
Collapse
|
16
|
Antimicrobial Activity of Rhenium Di- and Tricarbonyl Diimine Complexes: Insights on Membrane-Bound S. aureus Protein Binding. Pharmaceuticals (Basel) 2022; 15:ph15091107. [PMID: 36145328 PMCID: PMC9501577 DOI: 10.3390/ph15091107] [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: 08/04/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Antimicrobial resistance is one of the major human health threats, with significant impacts on the global economy. Antibiotics are becoming increasingly ineffective as drug-resistance spreads, imposing an urgent need for new and innovative antimicrobial agents. Metal complexes are an untapped source of antimicrobial potential. Rhenium complexes, amongst others, are particularly attractive due to their low in vivo toxicity and high antimicrobial activity, but little is known about their targets and mechanism of action. In this study, a series of rhenium di- and tricarbonyl diimine complexes were prepared and evaluated for their antimicrobial potential against eight different microorganisms comprising Gram-negative and -positive bacteria. Our data showed that none of the Re dicarbonyl or neutral tricarbonyl species have either bactericidal or bacteriostatic potential. In order to identify possible targets of the molecules, and thus possibly understand the observed differences in the antimicrobial efficacy of the molecules, we computationally evaluated the binding affinity of active and inactive complexes against structurally characterized membrane-bound S. aureus proteins. The computational analysis indicates two possible major targets for this class of compounds, namely lipoteichoic acids flippase (LtaA) and lipoprotein signal peptidase II (LspA). Our results, consistent with the published in vitro studies, will be useful for the future design of rhenium tricarbonyl diimine-based antibiotics.
Collapse
|
17
|
Sovari SN, Golding TM, Mbaba M, Mohunlal R, Egan TJ, Smith GS, Zobi F. Rhenium(I) derivatives of aminoquinoline and imidazolopiperidine-based ligands: Synthesis, in vitro and in silico biological evaluation against Plasmodium falciparum. J Inorg Biochem 2022; 234:111905. [PMID: 35752063 DOI: 10.1016/j.jinorgbio.2022.111905] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/23/2022] [Accepted: 06/12/2022] [Indexed: 12/31/2022]
Abstract
A small library of aminoquinoline and imidazolopiperidine (IMP)-based ligands, containing the 1,2,3-triazole moiety, and their corresponding tricarbonyl rhenium complexes were synthesised and their inhibitory activities evaluated against the chloroquine-sensitive (CQS) and multidrug-resistant (MDR) strains (NF54 and K1, respectively) of P. falciparum. The quinoline-based compounds (L1, L2, ReL1, and ReL2) were at least six-fold more potent than their IMP-based counterparts (L3, L4, ReL3, and ReL4) against both strains of P. falciparum, with the most promising compound (L1) displaying activity comparable to chloroquine diphosphate (CQDP) in the MDR strain. Additionally, all of the synthesised compounds have resistance indices less than CQDP. To gain insight into a possible mechanism of action, in silico hemozoin docking simulations were performed. These studies proposed that the tested compounds may act via hemozoin inhibition, as the new aminoquinoline-derivatives, with the exception of complex ReL2 (binding affinity: -12.62 kcal/mol), showed higher binding affinities than the reference drug chloroquine (CQ, -13.56 kcal/mol). Furthermore, the ligands exhibited superior binding affinity relative to their corresponding Re(I) complexes, which is reflected in their antiplasmodial activity.
Collapse
Affiliation(s)
- Sara Nasiri Sovari
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland
| | - Taryn M Golding
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Mziyanda Mbaba
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Roxanne Mohunlal
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7700, South Africa
| | - Gregory S Smith
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town 7700, South Africa.
| | - Fabio Zobi
- Department of Chemistry, Fribourg University, Chemin Du Musée 9, 1700 Fribourg, Switzerland.
| |
Collapse
|
18
|
Cooper SM, Siakalli C, White AJP, Frei A, Miller PW, Long NJ. Synthesis and anti-microbial activity of a new series of bis(diphosphine) rhenium(V) dioxo complexes. Dalton Trans 2022; 51:12791-12795. [PMID: 35920379 DOI: 10.1039/d2dt02157a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Rhenium-based metallodrugs have recently been highlighted as promising candidates for new antibiotics to combat multi-drug resistant (MDR) pathogens. A new class of rhenium(V) dioxo complexes were prepared from readily accessible diphosphine ligands, and have been shown to possess potent activity against Staphylococcus aureus (S. aureus) and Candida albicans (C. albicans) alongside low human cell toxicity.
Collapse
Affiliation(s)
- Saul M Cooper
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| | - Christina Siakalli
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| | - Andrew J P White
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| | - Angelo Frei
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| | - Philip W Miller
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| | - Nicholas J Long
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 82 Wood Lane, White City Campus, London, W12 0BZ, UK.
| |
Collapse
|
19
|
Lycosin-II Exhibits Antifungal Activity and Inhibits Dual-Species Biofilm by Candida albicans and Staphylococcus aureus. J Fungi (Basel) 2022; 8:jof8090901. [PMID: 36135626 PMCID: PMC9504746 DOI: 10.3390/jof8090901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The increase and dissemination of antimicrobial resistance is a global public health issue. To address this, new antimicrobial agents have been developed. Antimicrobial peptides (AMPs) exhibit a wide range of antimicrobial activities against pathogens, including bacteria and fungi. Lycosin-II, isolated from the venom of the spider Lycosa singoriensis, has shown antibacterial activity by disrupting membranes. However, the mode of action of Lycosin-II and its antifungal activity have not been clearly described. Therefore, we confirmed that Lycosin-II showed antifungal activity against Candida albicans (C. albicans). To investigate the mode of action, membrane-related assays were performed, including an evaluation of C. albicans membrane depolarization and membrane integrity after exposure to Lycosin-II. Our results indicated that Lycosin-II damaged the C. albicans membrane. Additionally, Lycosin-II induced oxidative stress through the generation of reactive oxygen species (ROS) in C. albicans. Moreover, Lycosin-II exhibited an inhibitory effect on dual-species biofilm formation by C. albicans and Staphylococcus aureus (S. aureus), which are the most co-isolated fungi and bacteria. These results revealed that Lycosin-II can be utilized against C. albicans and dual-species strain infections.
Collapse
|
20
|
Mendes SS, Marques J, Mesterházy E, Straetener J, Arts M, Pissarro T, Reginold J, Berscheid A, Bornikoel J, Kluj RM, Mayer C, Oesterhelt F, Friães S, Royo B, Schneider T, Brötz-Oesterhelt H, Romão CC, Saraiva LM. Synergetic Antimicrobial Activity and Mechanism of Clotrimazole-Linked CO-Releasing Molecules. ACS BIO & MED CHEM AU 2022; 2:419-436. [PMID: 35996473 PMCID: PMC9389576 DOI: 10.1021/acsbiomedchemau.2c00007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Several metal-based
carbon monoxide-releasing molecules (CORMs)
are active CO donors with established antibacterial activity. Among
them, CORM conjugates with azole antibiotics of type [Mn(CO)3(2,2′-bipyridyl)(azole)]+ display important synergies
against several microbes. We carried out a structure–activity
relationship study based upon the lead structure of [Mn(CO)3(Bpy)(Ctz)]+ by producing clotrimazole (Ctz) conjugates
with varying metal and ligands. We concluded that the nature of the
bidentate ligand strongly influences the bactericidal activity, with
the substitution of bipyridyl by small bicyclic ligands leading to
highly active clotrimazole conjugates. On the contrary, the metal
did not influence the activity. We found that conjugate [Re(CO)3(Bpy)(Ctz)]+ is more than the sum of its parts:
while precursor [Re(CO)3(Bpy)Br] has no antibacterial activity
and clotrimazole shows only moderate minimal inhibitory concentrations,
the potency of [Re(CO)3(Bpy)(Ctz)]+ is one order
of magnitude higher than that of clotrimazole, and the spectrum of
bacterial target species includes Gram-positive and Gram-negative
bacteria. The addition of [Re(CO)3(Bpy)(Ctz)]+ to Staphylococcus aureus causes a
general impact on the membrane topology, has inhibitory effects on
peptidoglycan biosynthesis, and affects energy functions. The mechanism
of action of this kind of CORM conjugates involves a sequence of events
initiated by membrane insertion, followed by membrane disorganization,
inhibition of peptidoglycan synthesis, CO release, and break down
of the membrane potential. These results suggest that conjugation
of CORMs to known antibiotics may produce useful structures with synergistic
effects that increase the conjugate’s activity relative to
that of the antibiotic alone.
Collapse
Affiliation(s)
- Sofia S Mendes
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Joana Marques
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Edit Mesterházy
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Jan Straetener
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Melina Arts
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Teresa Pissarro
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Jorgina Reginold
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Anne Berscheid
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Jan Bornikoel
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Robert M Kluj
- Institute of Microbiology and Infection Medicine, Dept. of Organismic Interactions, University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Christoph Mayer
- Institute of Microbiology and Infection Medicine, Dept. of Organismic Interactions, University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Filipp Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Sofia Friães
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Beatriz Royo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Tanja Schneider
- Institute for Pharmaceutical Microbiology, University of Bonn, University Clinic Bonn, Meckenheimer Allee 168, 53115 Bonn, Germany
| | - Heike Brötz-Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, Dept. of Microbial Bioactive Compounds, Cluster of Excellence Controlling Microbes to Fight Infection. University of Tuebingen, Auf der Morgenstelle 28, 72070 Tuebingen, Germany
| | - Carlos C Romão
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| | - Lígia M Saraiva
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República (EAN), 2780-157 Oeiras, Portugal
| |
Collapse
|
21
|
Suárez-Ortiz GA, Hernández-Correa R, Morales-Moreno MD, Toscano RA, Ramirez-Apan MT, Hernandez-Garcia A, Amézquita-Valencia M, Araiza-Olivera D. Diastereomeric Separation of Chiral fac-Tricarbonyl(iminopyridine) Rhenium(I) Complexes and Their Cytotoxicity Studies: Approach toward an Action Mechanism against Glioblastoma. J Med Chem 2022; 65:9281-9294. [PMID: 35776775 DOI: 10.1021/acs.jmedchem.2c00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of new (tricarbonyl)rhenium(I) complexes were synthesized using chiral bidentate ligands (+)/(-)-iminopyridines (LR/LS). The reaction yielded a mixture of mononuclear Re(I) diastereoisomers, formulated as fac-[Br(CO)3Re(S/R)L(S/R)]. Each single diastereoisomer was isolated and fully characterized. X-ray crystallography and circular dichroism spectra verified their enantiomeric nature. The cytotoxicity of each complex was evaluated against six cancer cell lines. The effect of the two complexes on viability, proliferation, and migration was analyzed on glioblastoma cell lines (U251 and LN229). Changes in the expression of histones, apoptotic, and key signaling proteins, as well as alterations in DNA structure, were also observed. These experiments showed that the chirality associated with both metal and ligand has a strong influence on cytotoxicity.
Collapse
Affiliation(s)
- Gloria A Suárez-Ortiz
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Rodrigo Hernández-Correa
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Melissa D Morales-Moreno
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Rubén A Toscano
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Maria Teresa Ramirez-Apan
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Armando Hernandez-Garcia
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Manuel Amézquita-Valencia
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| | - Daniela Araiza-Olivera
- Universidad Nacional Autónoma de México, Instituto de Química, Ciudad Universitaria, Ciudad de México 04510, México
| |
Collapse
|
22
|
Schutte-Smith M, Visser HG. Crystal and molecular structures of fac-[Re(Bid)(PPh 3)(CO) 3] [Bid is tropolone (TropH) and tribromotropolone (TropBr 3H)]. Acta Crystallogr C Struct Chem 2022; 78:351-359. [PMID: 35662135 PMCID: PMC9167630 DOI: 10.1107/s205322962200465x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
Two rhenium complexes, namely, fac-tricarbonyl(triphenylphosphane-κP)(tropolonato-κ2O,O')rhenium(I), [Re(C7H5O2)(C18H15P)(CO)3] or fac-[Re(Trop)(PPh3)(CO)3] (1), and fac-tricarbonyl(3,5,7-tribromotropolonato-κ2O,O')(triphenylphosphane-κP)rhenium(I), [Re(C7H2Br3O2)(C18H15P)(CO)3] or fac-[Re(TropBr3)(PPh3)(CO)3] (2) (TropH is tropolone and and TropBr3H is tribromotropolone), were synthesized and their crystal and molecular structures confirmed by single-crystal X-ray diffraction. Both crystallized in the space group P-1 and display an array of inter- and intramolecular interactions which were confirmed by solid-state 13C NMR spectroscopy using cross polarization magic angle spinning (CPMAS) techniques, as well as Hirshfeld surface analysis. The slightly longer Re-P distance of 1 [2.4987 (5) versus 2.4799 (11) Å for 1 and 2, respectively] suggests stronger back donation from the carbonyl groups in the former case, possibly due to the stronger electron-donating ability of the unsubstituted tropolonate ring system. However, this is not supported in the Re-CO bond distances of 1 and 2.
Collapse
Affiliation(s)
- Marietjie Schutte-Smith
- Department of Chemistry, University of the Free State, PO Box 339, Bloemfontein 9301, South Africa
| | - Hendrik Gideon Visser
- Department of Chemistry, University of the Free State, PO Box 339, Bloemfontein 9301, South Africa
| |
Collapse
|
23
|
Pavic A, Stojanovic Z, Pekmezovic M, Veljović Đ, O’Connor K, Malagurski I, Nikodinovic-Runic J. Polyenes in Medium Chain Length Polyhydroxyalkanoate (mcl-PHA) Biopolymer Microspheres with Reduced Toxicity and Improved Therapeutic Effect against Candida Infection in Zebrafish Model. Pharmaceutics 2022; 14:pharmaceutics14040696. [PMID: 35456530 PMCID: PMC9028145 DOI: 10.3390/pharmaceutics14040696] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 11/16/2022] Open
Abstract
Immobilizing antifungal polyenes such as nystatin (Nys) and amphotericin B (AmB) into biodegradable formulations is advantageous compared to free drug administration providing sustained release, reduced dosing due to localized targeting and overall reduced systemic drug toxicity. In this study, we encapsulated Nys and AmB in medium chain length polyhydroxyalkanoates (mcl-PHA) microspheres (7–8 µm in diameter). The obtained formulations have been validated for antifungal activity in vitro against a panel of pathogenic fungi including species of Candida, Aspergillus, Microsporum and Trichophyton genera and toxicity and efficacy in vivo using the zebrafish model of disseminated candidiasis. While free polyenes, especially AmB, were highly toxic to zebrafish embryos at the effective (MIC) doses, after their loading into mcl-PHA microspheres, inner organ toxicity and teratogenicity associated with both drugs were not observed, even at 100 × MIC doses. The obtained mcl-PHA/polyene formulations have successfully eradicated C. albicans infection and showed an improved therapeutic profile in zebrafish by enhancing infected embryos survival. This approach is contributing to the antifungal arsenal as polyenes, although the first broad-spectrum antifungals on the market are still the gold standard for treatment of fungal infections.
Collapse
Affiliation(s)
- Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (A.P.); (I.M.)
| | - Zoran Stojanovic
- Institute of Technical Sciences of the Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia;
| | - Marina Pekmezovic
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany;
| | - Đorđe Veljović
- Faculty of Technology and Metallurgy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Kevin O’Connor
- BiOrbic Bioeconomy SFI Research Centre, Belfield, D04 V1W Dublin, Ireland;
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, D04 V1W Dublin, Ireland
| | - Ivana Malagurski
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (A.P.); (I.M.)
| | - Jasmina Nikodinovic-Runic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia; (A.P.); (I.M.)
- Correspondence: ; Tel.: +381-11-397-6034
| |
Collapse
|
24
|
Radakovic N, Nikolić A, Jovanović NT, Stojković P, Stankovic N, Šolaja B, Opsenica I, Pavic A. Unraveling the anti-virulence potential and antifungal efficacy of 5-aminotetrazoles using the zebrafish model of disseminated candidiasis. Eur J Med Chem 2022; 230:114137. [PMID: 35077918 DOI: 10.1016/j.ejmech.2022.114137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 11/16/2022]
Abstract
Candida albicans remains the main causal agent of candidiasis, the most common fungal infection with disturbingly high mortality rates worldwide. The limited diversity and efficacy of clinical antifungal drugs, exacerbated by emerging drug resistance, have resulted in the failure of current antifungal therapies. This imposes an urgent demand for the development of innovative strategies for effective eradication of candidal infections. While the existing clinical drugs display fungicidal or fungistatic activity, the strategy specifically targeting C. albicans filamentation, as the most important virulence trait, represents an attractive approach for overcoming the drawbacks related to clinical antifungals. The results acquired in this study revealed the significant potential of 5-aminotetrazoles as a new class of effective and safe anti-virulence agents. Moreover, these novel agents were active when applied both alone and in combination with clinically approved polyenes. Complete prevention of C. albicans morphogenetic yeast-to-hyphae transition was achieved at doses as low as 1.3 μM under conditions mimicking various filamentation-responsive stimuli in the human body, while no cardio- or hepatotoxicity was observed at doses as high as 200 μM. The treatment of C. albicans-infected zebrafish embryos with nystatin alone had low efficacy, while the combination of nystatin and selected 5-aminotetrazoles prevented fungal filamentation, successfully eliminating the infection and rescuing the infected embryos from lethal disseminated candidiasis. In addition, the most potent anti-virulence 5-aminotetrazole prevented C. albicans in developing the resistance to nystatin when applied in combination, keeping the fungus sensitive to the antifungal drug.
Collapse
Affiliation(s)
- Natasa Radakovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade 152, Serbia
| | - Andrea Nikolić
- University of Belgrade - Faculty of Chemistry, PO Box 51, Studentski trg 16, 11158, Belgrade, Serbia
| | - Nataša Terzić Jovanović
- University of Belgrade - Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Njegoševa 12, 11000, Belgrade, Serbia
| | - Pavle Stojković
- University of Belgrade - Faculty of Chemistry, PO Box 51, Studentski trg 16, 11158, Belgrade, Serbia
| | - Nada Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade 152, Serbia
| | - Bogdan Šolaja
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000, Belgrade, Serbia
| | - Igor Opsenica
- University of Belgrade - Faculty of Chemistry, PO Box 51, Studentski trg 16, 11158, Belgrade, Serbia.
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade 152, Serbia.
| |
Collapse
|
25
|
Schindler K, Zobi F. Anticancer and Antibiotic Rhenium Tri- and Dicarbonyl Complexes: Current Research and Future Perspectives. Molecules 2022; 27:539. [PMID: 35056856 PMCID: PMC8777860 DOI: 10.3390/molecules27020539] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/23/2021] [Accepted: 01/12/2022] [Indexed: 12/20/2022] Open
Abstract
Organometallic compounds are increasingly recognized as promising anticancer and antibiotic drug candidates. Among the transition metal ions investigated for these purposes, rhenium occupies a special role. Its tri- and dicarbonyl complexes, in particular, attract continuous attention due to their relative ease of preparation, stability and unique photophysical and luminescent properties that allow the combination of diagnostic and therapeutic purposes, thereby permitting, e.g., molecules to be tracked within cells. In this review, we discuss the anticancer and antibiotic properties of rhenium tri- and dicarbonyl complexes described in the last seven years, mainly in terms of their structural variations and in vitro efficacy. Given the abundant literature available, the focus is initially directed on tricarbonyl complexes of rhenium. Dicarbonyl species of the metal ion, which are slowly gaining momentum, are discussed in the second part in terms of future perspective for the possible developments in the field.
Collapse
Affiliation(s)
| | - Fabio Zobi
- Department of Chemistry, Fribourg University, Chemin du Musée 9, 1700 Fribourg, Switzerland;
| |
Collapse
|
26
|
Herdman M, Werrett MV, Andrews PC. Aryl bismuth phosphinates [BiAr2(O(O)PRR’)]: structure-activity relationships for antibacterial activity and cytotoxicity. Dalton Trans 2022; 51:9323-9335. [DOI: 10.1039/d2dt00346e] [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
To study and evaluate the structure-activity relationships in di-aryl bismuth phosphinates on antibacterial activity and cytotoxicity a series of complexes containing ortho-methoxyphenyl, meta-methoxyphenyl, meta-tolyl and para-tolyl aryl groups; [Bi(o-MeOPh)2(O(O)P(H)Ph)]n 1,...
Collapse
|
27
|
Nolan VC, Rafols L, Harrison J, Soldevila-Barreda JJ, Crosatti M, Garton NJ, Wegrzyn M, Timms DL, Seaton CC, Sendron H, Azmanova M, Barry NP, Pitto-Barry A, Cox JA. Indole-containing arene-ruthenium complexes with broad spectrum activity against antibiotic-resistant bacteria. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100099. [PMID: 35059676 PMCID: PMC8760505 DOI: 10.1016/j.crmicr.2021.100099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022] Open
Abstract
A new family of indole-containing arene ruthenium organometallic compounds are active against several bacterial species and drug resistant strains Bactericidal activity observed against various Gram negative, Gram positive and acid-fast bacteria, demonstrating broad-spectrum inhibitory activity Compound series exhibits low toxicity against human cells Shows considerable promise as next generation antibiotics
Antimicrobial resistant (AMR) bacteria are emerging and spreading globally, threatening our ability to treat common infectious diseases. The development of new classes of antibiotics able to kill or inhibit the growth of such AMR bacteria through novel mechanisms of action is therefore urgently needed. Here, a new family of indole-containing arene ruthenium organometallic compounds are screened against several bacterial species and drug resistant strains. The most active complex [(p-cym)Ru(O-cyclohexyl-1H-indole-2-carbothioate)Cl] (3) shows growth inhibition and bactericidal activity against different organisms (Acinetobacter baumannii, Mycobacterium abscessus, Mycobacterium tuberculosis, Staphylococcus aureus, Salmonella enterica serovar Typhi and Escherichia coli), demonstrating broad-spectrum inhibitory activity. Importantly, this compound series exhibits low toxicity against human cells. Owing to the novelty of the antibiotic family, their moderate cytotoxicity, and their inhibitory activity against Gram positive, Gram negative and acid-fast, antibiotic resistant microorganisms, this series shows significant promise for further development.
Collapse
|