1
|
Rogala P, Jabłońska-Wawrzycka A, Czerwonka G, Hodorowicz M, Michałkiewicz S, Kalinowska-Tłuścik J, Karpiel M, Gałczyńska K. Ruthenium Complexes with Pyridazine Carboxylic Acid: Synthesis, Characterization, and Anti-Biofilm Activity. Molecules 2024; 29:5694. [PMID: 39683853 DOI: 10.3390/molecules29235694] [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: 10/30/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
As a result of drug resistance, many antimicrobial medicines become ineffective, making the infections more difficult to treat. Therefore, there is a need to develop new compounds with antibacterial activity. This role may be played, for example, by metal complexes with carboxylic acids. This study reports the formation and characterization of ruthenium complexes with pyridazine-3-carboxylic acid (pdz-3-COOH)-([(η6-p-cym)RuIICl(pdz-3-COO)] (1), [RuIIICl2(pdz-3-COO)2Na(H2O)]n(H2O)0.11 (2) and [RuIIICl2(pdz-3-COO)2Na(H2O)2]n (3). The synthesized compounds were analyzed using various spectroscopic and electrochemical techniques, with structure confirmation via SC-XRD analysis. Experimental data showed the ligand binds to metal ions bidentately through the nitrogen donor of the pyridazine ring and one carboxylate oxygen. To visualize intermolecular interactions, Hirshfeld surface analysis and 2D fingerprint plots were conducted. Furthermore, the impact of ruthenium compounds (1 and 2) on the planktonic growth of selected bacterial strains and the formation of Pseudomonas aeruginosa PAO1 biofilm was examined. Both complexes demonstrated comparable anti-biofilm activity and outperformed the free ligand. The effect of the complexes on selected virulence factors of P. aeruginosa PAO1 was also investigated. Compounds 1 and 2 show high suppressive activity in pyoverdine production, indicating that the virulence of the strain has been reduced. This inhibitory effect is similar to the inhibitory effect of ciprofloxacin. Within this context, the complexes exhibit promising antibacterial activities. Importantly, the compounds showed no cytotoxic effects on normal CHO-K1 cells. Additionally, a molecular docking approach and fluorescence spectroscopy were used to determine the interactions of ruthenium complexes with human serum albumin.
Collapse
Affiliation(s)
- Patrycja Rogala
- Institute of Chemistry, Jan Kochanowski University, 7 Uniwersytecka Str., 25-406 Kielce, Poland
| | | | - Grzegorz Czerwonka
- Institute of Biology, Jan Kochanowski University, 7 Uniwersytecka Str., 25-406 Kielce, Poland
| | - Maciej Hodorowicz
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Cracow, Poland
| | - Sławomir Michałkiewicz
- Institute of Chemistry, Jan Kochanowski University, 7 Uniwersytecka Str., 25-406 Kielce, Poland
| | | | - Marta Karpiel
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Cracow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 11 Lojasiewicza Str., 30-348 Cracow, Poland
| | - Katarzyna Gałczyńska
- Institute of Biology, Jan Kochanowski University, 7 Uniwersytecka Str., 25-406 Kielce, Poland
| |
Collapse
|
2
|
Deng W, Xue RY, Xiao SX, Wang JT, Liao XW, Yu RJ, Xiong YS. Discovery of quaternized pyridine-thiazole-ruthenium complexes as potent anti-Staphylococcus aureus agents. Eur J Med Chem 2024; 277:116712. [PMID: 39106657 DOI: 10.1016/j.ejmech.2024.116712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 08/09/2024]
Abstract
Quaternization of ruthenium complexes may be a promising strategy for the development of new antibiotics. In response to the increasing bacterial resistance, we integrated the quaternary amine structure into the design of ruthenium complexes and evaluated their antibacterial activity. All the ruthenium complexes showed good antibacterial activity against the tested Staphylococcus aureus (S. aureus). Ru-8 was the most effective antibacterial agent that displayed excellent antibacterial activity against S. aureus (MIC = 0.78-1.56 μg/mL). In vitro experiments showed that all nine ruthenium complexes had low hemolytic toxicity to rabbit erythrocytes. Notably, Ru-8 was found to disrupt bacterial cell membranes, alter their permeability, and induce ROS production in bacteria, all the above leading to the death of bacteria without inducing drug resistance. To further explore the antibacterial activity of Ru-8in vivo, we established a mouse skin wound infection model and a G. mellonella larvae infection model. Ru-8 exhibited significant antibacterial efficacy against S. aureus in vivo and low toxicity to mouse tissues. The Ru-8 showed low toxicity to Raw264.7 cells (mouse monocyte macrophage leukemia cells). This study indicates that the ruthenium complex ruthenium quaternary was a promising strategy for the development of new antibacterial agents.
Collapse
Affiliation(s)
- Wei Deng
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Run-Yu Xue
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Su-Xin Xiao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Jin-Tao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Ru-Jian Yu
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
3
|
Zahirović A, Fetahović S, Feizi-Dehnayebi M, Višnjevac A, Bešta-Gajević R, Kozarić A, Martić L, Topčagić A, Roca S. Dual Antimicrobial-Anticancer Potential, Hydrolysis, and DNA/BSA Binding Affinity of a Novel Water-Soluble Ruthenium-Arene Ethylenediamine Schiff base (RAES) Organometallic. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 318:124528. [PMID: 38801789 DOI: 10.1016/j.saa.2024.124528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The need for a systematic approach in developing new metal-based drugs with dual anticancer-antimicrobial properties is emphasized by the vulnerability of cancer patients to bacterial infections. In this context, a novel organometallic assembly was designed, featuring ruthenium(II) coordination with p-cymene, one chlorido ligand, and a bidentate neutral Schiff base derived from 4-methoxybenzaldehyde and N,N-dimethylethylenediamine. The compound was extensively characterized in both solid-state and solution, employing single crystal X-ray diffraction, nuclear magnetic resonance, infrared, ultraviolet-visible spectroscopy, and density functional theory, alongside Hirshfeld surface analysis. The hydrolysis kinetic was thoroughly investigated, revealing the important role of the chloro-aqua equilibrium in the dynamics of binding with deoxyribonucleic acid and bovine serum albumin. Notably, the aqua species exhibited a pronounced affinity for deoxyribonucleic acid, engaging through electrostatic and hydrogen bonding interactions, while the chloro species demonstrated groove-binding properties. Interaction with albumin revealed distinct binding mechanisms. The aqua species displayed covalent binding, contrasting with the ligand-like van der Waals interactions and hydrogen bonding observed with the chloro specie. Molecular docking studies highlighted site-specific interactions with biomolecular targets. Remarkably, the compound exhibited wide spectrum moderate antimicrobial activity against Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans, coupled with low micromolar cytotoxic activity against human colorectal adenocarcinoma cells and significant activity against human leukemic monocyte lymphoma cells. The presented findings encourage further development of this compound, promising avenues for its evolution into a versatile therapeutic agent targeting both infectious diseases and cancer.
Collapse
Affiliation(s)
- Adnan Zahirović
- Laboratory for Inorganic and Bioinorganic Chemistry, Department of Chemistry, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Selma Fetahović
- Laboratory for Inorganic and Bioinorganic Chemistry, Department of Chemistry, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Aleksandar Višnjevac
- Laboratory for Chemical and Biological Crystallography, Division of Physical Chemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Renata Bešta-Gajević
- Department of Biology, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Amina Kozarić
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Lora Martić
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
| | - Anela Topčagić
- Laboratory for Inorganic and Bioinorganic Chemistry, Department of Chemistry, Faculty of Science, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Sunčica Roca
- NMR Centre, Ruđer Bošković Institute, Zagreb, Croatia.
| |
Collapse
|
4
|
Alven S, Gandidzanwa S, Ngalo B, Poswayo O, Madanhire T, Aderibigbe BA, Tshentu Z. Platinum Group Metals Nanoparticles in Breast Cancer Therapy. Pharmaceutics 2024; 16:1162. [PMID: 39339199 PMCID: PMC11434984 DOI: 10.3390/pharmaceutics16091162] [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: 07/19/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Despite various methods currently used in cancer therapy, breast cancer remains the leading cause of morbidity and mortality worldwide. Current therapeutics face limitations such as multidrug resistance, drug toxicity and off-target effects, poor drug bioavailability and biocompatibility, and inefficient drug delivery. Nanotechnology has emerged as a promising approach to cancer diagnosis, imaging, and therapy. Several preclinical studies have demonstrated that compounds and nanoparticles formulated from platinum group metals (PGMs) effectively treat breast cancer. PGMs are chemically stable, easy to functionalise, versatile, and tunable. They can target hypoxic microenvironments, catalyse the production of reactive oxygen species, and offer the potential for combination therapy. PGM nanoparticles can be incorporated with anticancer drugs to improve efficacy and can be attached to targeting moieties to enhance tumour-targeting efficiency. This review focuses on the therapeutic outcomes of platinum group metal nanoparticles (PGMNs) against various breast cancer cells and briefly discusses clinical trials of these nanoparticles in breast cancer treatment. It further illustrates the potential applications of PGMNs in breast cancer and presents opportunities for future PGM-based nanomaterial applications in combatting breast cancer.
Collapse
Affiliation(s)
- Sibusiso Alven
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| | | | - Basabele Ngalo
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Olwethu Poswayo
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| | - Tatenda Madanhire
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
- Department of Chemistry, University of South Africa, Johannesburg 1710, South Africa
| | | | - Zenixole Tshentu
- Department of Chemistry, Nelson Mandela University, Gqeberha 6001, South Africa
| |
Collapse
|
5
|
Žužek MC. Advances in Cholinesterase Inhibitor Research-An Overview of Preclinical Studies of Selected Organoruthenium(II) Complexes. Int J Mol Sci 2024; 25:9049. [PMID: 39201735 PMCID: PMC11354293 DOI: 10.3390/ijms25169049] [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: 07/11/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Cholinesterase (ChE) inhibitors are crucial therapeutic agents for the symptomatic treatment of certain chronic neurodegenerative diseases linked to functional disorders of the cholinergic system. Significant research efforts have been made to develop novel derivatives of classical ChE inhibitors and ChE inhibitors with novel scaffolds. Over the past decade, ruthenium complexes have emerged as promising novel therapeutic alternatives for the treatment of neurodegenerative diseases. Our research group has investigated a number of newly synthesized organoruthenium(II) complexes for their inhibitory activity against acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). Three complexes (C1a, C1-C, and C1) inhibit ChE in a pharmacologically relevant range. C1a reversibly inhibits AChE and BChE without undesirable peripheral effects, making it a promising candidate for the treatment of Alzheimer's disease. C1-Cl complex reversibly and competitively inhibits ChEs, particularly AChE. It inhibits nerve-evoked skeletal muscle twitch and tetanic contraction in a concentration-dependent manner with no effect on directly elicited twitch and tetanic contraction and is promising for further preclinical studies as a competitive neuromuscular blocking agent. C1 is a selective, competitive, and reversible inhibitor of BChE that inhibits horse serum BChE (hsBChE) without significant effect on the peripheral neuromuscular system and is a highly species-specific inhibitor of hsBChE that could serve as a species-specific drug target. This research contributes to the expanding knowledge of ChE inhibitors based on ruthenium complexes and highlights their potential as promising therapeutic candidates for chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Monika C Žužek
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
Vadakkedathu Palakkeezhillam VN, Haribabu J, Kumar VS, Manakkadan V, Rasin P, Muena JP, Dharmasivam M, Sreekanth A. Biomolecular Interactions and Anticancer Mechanisms of Ru(II)-Arene Complexes of Cinnamaldehyde-Derived Thiosemicarbazone Ligands: Analysis Combining In Silico and In Vitro Approaches. ACS APPLIED BIO MATERIALS 2024; 7:5622-5639. [PMID: 39087675 DOI: 10.1021/acsabm.4c00689] [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: 08/02/2024]
Abstract
Our study focuses on synthesizing and exploring the potential of three N-(4) substituted thiosemicarbazones derived from cinnamic aldehyde, alongside their Ru(II)-(η6 -p-cymene)/(η6-benzene) complexes. The synthesized compounds were comprehensively characterized using a range of analytical techniques, including FT-IR, UV-visible spectroscopy, NMR (1H, 13C), and HRMS. We investigated their electronic and physicochemical properties via density functional theory (DFT). X-ray crystal structures validated structural differences identified by DFT. Molecular docking predicted promising bioactivities, supported by experimental observations. Notably, docking with EGFR suggested an inhibitory potential against this cancer-related protein. Spectroscopic titrations revealed significant DNA/BSA binding affinities, particularly with DNA intercalation and BSA hydrophobic interactions. RuPCAM displayed the strongest binding affinity with DNA (Kb = 6.23 × 107 M-1) and BSA (Kb = 9.75 × 105 M-1). Assessed the cytotoxicity of the complexes on cervical cancer cells (HeLa), and breast cancer cells (MCF-7 and MDA-MB 231), revealing remarkable potency. Additionally, selectivity was assessed by examining MCF-10a normal cell lines. The active complexes were found to trigger apoptosis, a vital cellular process crucial for evaluating their potential as anticancer agents utilizing staining assays and flow cytometry analysis. Intriguingly, complexation with Ru(II)-arene precursors significantly amplified the bioactivity of thiosemicarbazones, unveiling promising avenues toward the creation of powerful anticancer agents.
Collapse
Affiliation(s)
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, Copiapo 1532502, Chile
| | - Vaishnu Suresh Kumar
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu, India-620015
| | - Vipin Manakkadan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu, India-620015
| | - Puthiyavalappil Rasin
- Centre for Nonlinear Systems, Chennai Institute of Technology (CIT), Chennai 600069, India
| | - Juan Pablo Muena
- Departmento de Quimica y Biologia, Facultad de Ciencias Naturales, Universidad de Atacama, Av. Copiapo 485, Copiapo 1530000, Chile
| | - Mahendiran Dharmasivam
- Department of Chemistry, Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4222, Australia
| | - Anandaram Sreekanth
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu, India-620015
| |
Collapse
|
7
|
Boubaker G, Bernal A, Vigneswaran A, Imhof D, de Sousa MCF, Hänggeli KPA, Haudenschild N, Furrer J, Păunescu E, Desiatkina O, Hemphill A. In vitro and in vivo activities of a trithiolato-diRuthenium complex conjugated with sulfadoxine against the apicomplexan parasite Toxoplasma gondii. Int J Parasitol Drugs Drug Resist 2024; 25:100544. [PMID: 38703737 PMCID: PMC11087982 DOI: 10.1016/j.ijpddr.2024.100544] [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: 01/29/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
Organometallic compounds, including Ruthenium complexes, have been widely developed as anti-cancer chemotherapeutics, but have also attracted much interest as potential anti-parasitic drugs. Recently hybrid drugs composed of organometallic Ruthenium moieties that were complexed to different antimicrobial agents were synthesized. One of these compounds, a trithiolato-diRuthenium complex (RU) conjugated to sulfadoxine (SDX), inhibited proliferation of Toxoplasma gondii tachyzoites grown in human foreskin fibroblast (HFF) monolayers with an IC50 < 150 nM, while SDX and the non-modified RU complex applied either individually or as an equimolar mixture were much less potent. In addition, conjugation of SDX to RU lead to decreased HFF cytotoxicity. RU-SDX did not impair the in vitro proliferation of murine splenocytes at concentrations ranging from 0.1 to 0.5 μM but had an impact at 2 μM, and induced zebrafish embryotoxicity at 20 μM, but not at 2 or 0.2 μM. RU-SDX acted parasitostatic but not parasiticidal, and induced transient ultrastructural changes in the mitochondrial matrix of tachyzoites early during treatment. While other compounds that target the mitochondrion such as the uncouplers FCCP and CCCP and another trithiolato-Ruthenium complex conjugated to adenine affected the mitochondrial membrane potential, no such effect was detected for RU-SDX. Evaluation of the in vivo efficacy of RU-SDX in a murine T. gondii oocyst infection model comprised of non-pregnant outbred CD1 mice showed no effects on the cerebral parasite burden, but reduced parasite load in the eyes and in heart tissue.
Collapse
Affiliation(s)
- Ghalia Boubaker
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Alice Bernal
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Anitha Vigneswaran
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Dennis Imhof
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Maria Cristina Ferreira de Sousa
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland.
| | - Kai Pascal Alexander Hänggeli
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland.
| | - Noé Haudenschild
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| | - Emilia Păunescu
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| | - Oksana Desiatkina
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| | - Andrew Hemphill
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern. Länggass-Strasse 122, 3012, Bern, Switzerland.
| |
Collapse
|
8
|
Genna V, Reyes-Fraile L, Iglesias-Fernandez J, Orozco M. Nucleic acids in modern molecular therapies: A realm of opportunities for strategic drug design. Curr Opin Struct Biol 2024; 87:102838. [PMID: 38759298 DOI: 10.1016/j.sbi.2024.102838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/19/2024]
Abstract
RNA vaccines have made evident to society what was already known by the scientific community: nucleic acids will be the "drugs of the future." By modifying the genome, interfering in transcription or translation, and by introducing new catalysts into the cell or by mimicking antibody effects, nucleic acids can generate therapeutic activities that are not accessible by any other therapeutic agents. There are, however, challenges that need to be solved in the next few years to make nucleic acids usable in a wide range of therapeutic scenarios. This review illustrates how simulation methods can help achieve this goal.
Collapse
Affiliation(s)
- Vito Genna
- NBD|Nostrum Biodiscovery, Josep Tarradellas 8-10, Barcelona 08019, Spain. https://twitter.com/_VitoGenna_
| | - Laura Reyes-Fraile
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain; Sixfold Bioscience Ltd, Translational & Innovation Hub, 84 Wood Ln, London W12 0BZ, United Kingdom
| | | | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, Barcelona 08028, Spain; Department of Biochemistry and Biomedicine, University of Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
9
|
Es-Sounni B, Harboul K, Mouhib A, Alanazi AS, Hefnawy M, Bakhouch M, Benali T, Hammani K, Mazoir N, El Yazidi M, Benharref A, Fahim M. Ruthenium(II) Complex-Based Tetradentate Schiff Bases: Synthesis, Spectroscopic, Antioxidant, and Antibacterial Investigations. Int J Mol Sci 2024; 25:7879. [PMID: 39063120 PMCID: PMC11277530 DOI: 10.3390/ijms25147879] [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: 06/01/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
In this work, we describe the synthesis of novel Ruthenium (II) complex-based salen Schiff bases. The obtained Ruthenium (II) complexes are characterized using usual spectroscopic and spectrometric techniques, viz., IR, UV-Vis, NMR (1H and 13C), powder X-ray diffraction, and HRMS. Further techniques, such as DTA-TGA and elemental analysis, are used to well establish the structure of the obtained complexes. Octahedral geometries are tentatively proposed for the new Ru(II) complexes. The measured molar conductance for the Ruthenium (II) complexes shows their electrolytic nature (4.24-4.44 S/m). The new Ru(II) complexes are evaluated for their antioxidant and antibacterial activities. The DPPH radical scavenging, FRAP, and total antioxidant capacity (TAC) assays show that the obtained complexes are more potent than the used positive control. They also exhibit promising antibacterial responses against pathogen bacteria: [RuH2L3Cl2] exhibits an important inhibition against Bacillus subtilis DSM 6633, with an inhibition zone of 21 ± 1.41 mm with an MIC value of 0.39 mg/mL, and Proteus mirabilis INH, with 16.50 ± 0.70 mm and an MIC value of 0.78 mg/mL, while [RuH2L2Cl2] exerts interesting antibacterial effects versus Bacillus subtilis DSM 6633 (21 ± 1.41 mm) and Proteus mirabilis INH (25.5 ± 0.70 mm) with equal MIC values of 0.97 mg/mL.
Collapse
Affiliation(s)
- Bouchra Es-Sounni
- Laboratory of Innovative Materials and Biotechnology of Naturel Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco; (B.E.-S.)
| | - Kaoutar Harboul
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
| | - Ayoub Mouhib
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Ashwag S. Alanazi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh 1167, Saudi Arabia;
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mohamed Bakhouch
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Taoufiq Benali
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Safi 46000, Morocco
| | - Khalil Hammani
- Laboratory of Natural Resources and Environment, Polydisciplinary Faculty of Taza, Sidi Mohamed Ben Abdellah University of Fez, Taza 30050, Morocco (T.B.); (K.H.)
| | - Noureddine Mazoir
- Bioorganic Chemistry Team, Department of Chemistry, Faculty of Sciences, Chouaïb Doukkali University, El Jadida 24000, Morocco
| | - Mohamed El Yazidi
- Engineering Laboratory of Organometallic and Molecular Materials and Environment, Faculty of Sciences Dhar El Mahraz, University Sidi Mohamed Ben Abdellah, Fez 30000, Morocco;
| | - Ahmed Benharref
- Laboratory of Natural Substances Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco;
| | - Mohammed Fahim
- Laboratory of Innovative Materials and Biotechnology of Naturel Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco; (B.E.-S.)
| |
Collapse
|
10
|
Bishara Robertson IL, Zhang H, Reisner E, Butt JN, Jeuken LJC. Engineering of bespoke photosensitiser-microbe interfaces for enhanced semi-artificial photosynthesis. Chem Sci 2024; 15:9893-9914. [PMID: 38966358 PMCID: PMC11220614 DOI: 10.1039/d4sc00864b] [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: 02/04/2024] [Accepted: 05/20/2024] [Indexed: 07/06/2024] Open
Abstract
Biohybrid systems for solar fuel production integrate artificial light-harvesting materials with biological catalysts such as microbes. In this perspective, we discuss the rational design of the abiotic-biotic interface in biohybrid systems by reviewing microbes and synthetic light-harvesting materials, as well as presenting various approaches to coupling these two components together. To maximise performance and scalability of such semi-artificial systems, we emphasise that the interfacial design requires consideration of two important aspects: attachment and electron transfer. It is our perspective that rational design of this photosensitiser-microbe interface is required for scalable solar fuel production. The design and assembly of a biohybrid with a well-defined electron transfer pathway allows mechanistic characterisation and optimisation for maximum efficiency. Introduction of additional catalysts to the system can close the redox cycle, omitting the need for sacrificial electron donors. Studies that electronically couple light-harvesters to well-defined biological entities, such as emerging photosensitiser-enzyme hybrids, provide valuable knowledge for the strategic design of whole-cell biohybrids. Exploring the interactions between light-harvesters and redox proteins can guide coupling strategies when translated into larger, more complex microbial systems.
Collapse
Affiliation(s)
| | - Huijie Zhang
- Leiden Institute of Chemistry, Leiden University PO Box 9502 Leiden 2300 RA the Netherlands
| | - Erwin Reisner
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW UK
| | - Julea N Butt
- School of Chemistry and School of Biological Sciences, University of East Anglia Norwich Research Park Norwich NR4 7TJ UK
| | - Lars J C Jeuken
- Leiden Institute of Chemistry, Leiden University PO Box 9502 Leiden 2300 RA the Netherlands
| |
Collapse
|
11
|
Chandra A, Basu P, Raha S, Dhibar P, Bhattacharya S. Development of ruthenium complexes with S-donor ligands for application in synthesis, catalytic acceptorless alcohol dehydrogenation and crossed-aldol condensation. Dalton Trans 2024; 53:10675-10685. [PMID: 38860941 DOI: 10.1039/d4dt00985a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The reaction of [Ru(dmso)4Cl2] with a potassium salt of four xanthate (RO-C(S)S-; R = Me, Et, iPr and tBu) ligands (depicted as Ln; n = 1-4) in hot methanol afforded a group of mixed-ligand complexes of type [Ru(Ln)2(dmso)2]. The crystal structures of all the four complexes have been determined, which show that the xanthate ligands are bound to the metal center forming four-membered chelates and dmso is coordinated through sulfur and they are mutually cis. The relative thermodynamic stability of this cis and the other possible trans-isomers of these complexes has been assessed with the help of DFT calculations, which have revealed that the cis-isomer is the more stable isomer. The coordinated dmso in the [Ru(Ln)2(dmso)2] complexes could be easily displaced by chelating bidentate ligands (depicted as L') to furnish complexes of type [Ru(Ln)2(L')], as demonstrated through isolation of two such complexes, viz. [Ru(L3)2(bpy)] and [Ru(L2)2(phen)] (bpy = 2,2'-bipyridine and phen = 1,10-phenanthroline). The crystal structure of [Ru(L3)2(bpy)] has been determined and the structure of [Ru(L2)2(phen)] has been optimized by the DFT method. The electronic spectra of the four [Ru(Ln)2(dmso)2] complexes and the two derivatives ([Ru(Ln)2(L')]; n = 3, L' = bpy; n = 2, L' = phen), recorded in dichloromethane solutions, show intense absorptions spanning the visible and ultraviolet regions, which have been analyzed by the TDDFT method. The [Ru(Ln)2(dmso)2] complexes are found to serve as efficient catalyst precursors for the acceptorless dehydrogenation of 2-propanol followed by crossed-aldol condensation with substituted benzaldehydes (and related aldehydes), using tert-butoxide as the co-catalyst, producing dibenzylideneacetone derivatives in good yields.
Collapse
Affiliation(s)
- Anushri Chandra
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata - 700032, India.
| | - Pousali Basu
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata - 700032, India.
| | - Shreya Raha
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata - 700032, India.
| | - Papu Dhibar
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata - 700032, India.
- Department of Chemistry, Brainware University, Kolkata 700 125, India
| | - Samaresh Bhattacharya
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata - 700032, India.
| |
Collapse
|
12
|
Deng W, Zhang CY, Dou LX, Huang LT, Wang JT, Liao XW, Wang LP, Yu RJ, Xiong YS. Polypyridyl ruthenium complexes with benzothiazole moiety as membrane disruptors and anti-resistance agents for Staphylococcus aureus. J Inorg Biochem 2024; 254:112517. [PMID: 38460482 DOI: 10.1016/j.jinorgbio.2024.112517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Developing new antimicrobials to combat drug-resistant bacterial infections is necessary due to the increasing problem of bacterial resistance. In this study, four metallic ruthenium complexes modified with benzothiazoles were designed, synthesized and subjected to bio-evaluated. Among them, Ru-2 displayed remarkable inhibitory activity against Staphylococcus aureus (S. aureus) with a minimum inhibitory concentration (MIC) of 1.56 μg/mL. Additionally, it showcased low hemolytic toxicity (HC50 > 200 μg/mL) and the ability to effectively eradicate S. aureus without fostering drug resistance. Further investigation into the antibacterial mechanism suggested that Ru-2 may target the phospholipid component of S. aureus, leading to the disruption of the bacterial cell membrane and subsequent leakage of cell contents (nucleic acid, protein, and ONPG), ultimately resulting in the death of the bacterial cell. In vivo studies, both the G. mellonella larvae and the mouse skin infection models were conducted, indicated that Ru-2 could potentially serve as a viable candidate for the treatment of S. aureus infection. It exhibited no toxic or side effects on normal tissues. The results suggest that benzothiazole-modified ruthenium complexes may have potential as membrane-active antimicrobials against drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Wei Deng
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Chun-Yan Zhang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Li-Xin Dou
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Li-Ting Huang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Jin-Tiao Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Xiang-Wen Liao
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Li-Ping Wang
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Ru-Jian Yu
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Yan-Shi Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
13
|
Ng XY, Fong KW, Kiew LV, Chung PY, Liew YK, Delsuc N, Zulkefeli M, Low ML. Ruthenium(II) polypyridyl complexes as emerging photosensitisers for antibacterial photodynamic therapy. J Inorg Biochem 2024; 250:112425. [PMID: 37977020 DOI: 10.1016/j.jinorgbio.2023.112425] [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: 09/18/2023] [Revised: 11/01/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Photodynamic therapy (PDT) has recently emerged as a potential valuable alternative to treat microbial infections. In PDT, singlet oxygen is generated in the presence of photosensitisers and oxygen under light irradiation of a specific wavelength, causing cytotoxic damage to bacteria. This review highlights different generations of photosensitisers and the common characteristics of ideal photosensitisers. It also focuses on the emergence of ruthenium and more specifically on Ru(II) polypyridyl complexes as metal-based photosensitisers used in antimicrobial photodynamic therapy (aPDT). Their photochemical and photophysical properties as well as structures are discussed while relating them to their phototoxicity. The use of Ru(II) complexes with recent advancements such as nanoformulations, combinatory therapy and photothermal therapy to improve on previous shortcomings of the complexes are outlined. Future perspectives of these complexes used in two-photon PDT, photoacoustic imaging and sonotherapy are also discussed. This review covers the literature published from 2017 to 2023.
Collapse
Affiliation(s)
- Xiao Ying Ng
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Kar Wai Fong
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan, Republic of China
| | - Pooi Yin Chung
- Department of Microbiology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Yun Khoon Liew
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicolas Delsuc
- Laboratoire des Biomolécules, LBM, Département de Chimie, École Normale Supérieur, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Mohd Zulkefeli
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia.
| | - May Lee Low
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia.
| |
Collapse
|
14
|
D’Amato A, Mariconda A, Iacopetta D, Ceramella J, Catalano A, Sinicropi MS, Longo P. Complexes of Ruthenium(II) as Promising Dual-Active Agents against Cancer and Viral Infections. Pharmaceuticals (Basel) 2023; 16:1729. [PMID: 38139855 PMCID: PMC10747139 DOI: 10.3390/ph16121729] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Poor responses to medical care and the failure of pharmacological treatment for many high-frequency diseases, such as cancer and viral infections, have been widely documented. In this context, numerous metal-based substances, including cisplatin, auranofin, various gold metallodrugs, and ruthenium complexes, are under study as possible anticancer and antiviral agents. The two Ru(III) and Ru(II) complexes, namely, BOLD-100 and RAPTA-C, are presently being studied in a clinical trial and preclinical studies evaluation, respectively, as anticancer agents. Interestingly, BOLD-100 has also recently demonstrated antiviral activity against SARS-CoV-2, which is the virus responsible for the COVID-19 pandemic. Over the last years, much effort has been dedicated to discovering new dual anticancer-antiviral agents. Ru-based complexes could be very suitable in this respect. Thus, this review focuses on the most recent studies regarding newly synthesized Ru(II) complexes for use as anticancer and/or antiviral agents.
Collapse
Affiliation(s)
- Assunta D’Amato
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| | | | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| |
Collapse
|
15
|
Teixeira RG, Mészáros JP, Matos B, Côrte-Real L, Xavier CPR, Fontrodona X, Garcia MH, Romero I, Spengler G, Vasconcelos MH, Tomaz AI, Enyedy ÉA, Valente A. Novel family of [RuCp(N,N)(P)] + compounds with simultaneous anticancer and antibacterial activity: Biological evaluation and solution chemistry studies. Eur J Med Chem 2023; 262:115922. [PMID: 37944388 DOI: 10.1016/j.ejmech.2023.115922] [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: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
A family of ten novel ruthenium(II)-cyclopentadienyl organometallics of general formula [Ru(η5-C5H5)(N,N)(PPh2(C6H4COOR)][CF3SO3] (1-10) in which (N,N) = 4,4'-R'-2,2'-bipyridyl (R = -H or -CH2CH2OH; R' = -H, -CH3, -OCH3, -CH2OH, and -CH2-biotin) was prepared from [Ru(η5-C5H5)(PPh2(C6H4COOH))2Cl]. All compounds were fully characterized by means of several spectroscopic and analytical techniques, and the molecular structures of [Ru(η5-C5H5)(PPh2(C6H4COOH))2Cl], 1, 3 and 4 have been additionally studied by single-crystal X-ray diffraction. The anticancer activity of all compounds was evaluated in sensitive and multidrug-resistant counterpart cell lines from human colorectal cancer (Colo 205 and Colo 320) and non-small cell lung cancer NSCLC (A549, NCI-H460 versus NCI-H460/R) as well. Notably, compounds 6 and 7 (R CH2CH2OH and (N,N) = bipy or Me2bipy, respectively) showed antiproliferative effect against both cell lines with high intrinsic selectivity towards cancer cells. The antibacterial activity of all compounds was also evaluated against both Gram negative and Gram positive strains, and some compounds in the series showed potent antibacterial activity against Staphylococcus aureus strains, including the methicillin-resistant MRSA strains. Solution speciation studies revealed that the complexes bearing the PPh2(C6H4COO-) ligand are neutral at physiological pH (7.4) in contrast with their ethylene glycol derivatives that have a permanent positive charge. While all compounds are lipophilic, the difference in the distribution coefficient for neutral and charged complexes is around one order of magnitude. Complexes 6 and 7 exhibited excellent biological activity and were selected for further studies. Spectrofluorometric methods were used to investigate their interaction with biomolecules such as human serum albumin (HSA) and calf thymus DNA (ct-DNA). For these complexes, binding site II of HSA is a possible binding pocket through non-covalent interactions. The release of ethidium from the DNA adduct by the charged complexes proves their interaction with DNA in contrast to the neutral ones. In conclusion, Ru(II)-cyclopentadienyl complexes with 2,2'-bipyridyl-derivatives and an ethylene glycol moiety tethered to the phenylphosphane co-ligand are very promising from a therapeutic perspective, in particular complexes 6 and 7 that display remarkable antibacterial activity with a high anti-proliferative effect against colon and non-small cell lung cancers, both clinically challenging neoplasias in need of effective solutions.
Collapse
Affiliation(s)
- Ricardo G Teixeira
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - János P Mészáros
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary
| | - Beatriz Matos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal
| | - Leonor Côrte-Real
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Cristina P R Xavier
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal
| | - Xavier Fontrodona
- Departament de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/ M. Aurèlia Campmany, 69, E-17003, Girona, Spain
| | - M Helena Garcia
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Isabel Romero
- Departament de Química and Serveis Tècnics de Recerca, Universitat de Girona, C/ M. Aurèlia Campmany, 69, E-17003, Girona, Spain
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, 6725, Szeged, Hungary
| | - M Helena Vasconcelos
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, 4200-135, Porto, Portugal; Department of Biological Sciences, Faculty of Pharmacy of the University of Porto (FFUP), Porto, Portugal
| | - Ana Isabel Tomaz
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Éva A Enyedy
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary; MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, H-6720, Szeged, Hungary
| | - Andreia Valente
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal.
| |
Collapse
|
16
|
Bugnon Q, Melendez C, Desiatkina O, Fayolles de Chaptes L, Holzer I, Păunescu E, Hilty M, Furrer J. In vitro antibacterial activity of dinuclear thiolato-bridged ruthenium(II)-arene compounds. Microbiol Spectr 2023; 11:e0095423. [PMID: 37815336 PMCID: PMC10714934 DOI: 10.1128/spectrum.00954-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/28/2023] [Indexed: 10/11/2023] Open
Abstract
IMPORTANCE The in vitro assessment of diruthenium(II)-arene compounds against Escherichia coli, Streptococcus pneumoniae, and Staphylococcus aureus showed a significant antibacterial activity of some compounds against S. pneumoniae, with minimum inhibitory concentration (MIC) values ranging from 1.3 to 2.6 µM, and a medium activity against E. coli, with MIC of 25 µM. The nature of the substituents anchored on the bridging thiols and the compounds molecular weight appear to significantly influence the antibacterial activity. Fluorescence microscopy showed that these ruthenium compounds enter the bacteria and do not accumulate in the cell wall of gram-positive bacteria. These diruthenium(II)-arene compounds exhibit promising activity against S. aureus and S. pneumoniae and deserve to be considered for further studies, especially the compounds bearing larger benzo-fused lactam substituents.
Collapse
Affiliation(s)
- Quentin Bugnon
- Department of Medicine, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Camilo Melendez
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Oksana Desiatkina
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Louis Fayolles de Chaptes
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Isabelle Holzer
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Emilia Păunescu
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| | - Markus Hilty
- Department of Medicine, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceuticals Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Youf R, Ghanem R, Nasir A, Lemercier G, Montier T, Le Gall T. Impact of mucus and biofilm on antimicrobial photodynamic therapy: Evaluation using Ruthenium(II) complexes. Biofilm 2023; 5:100113. [PMID: 37396462 PMCID: PMC10313506 DOI: 10.1016/j.bioflm.2023.100113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 07/04/2023] Open
Abstract
The biofilm lifestyle of bacterial pathogens is a hallmark of chronic lung infections such as in cystic fibrosis (CF) patients. Bacterial adaptation to the complex conditions in CF-affected lungs and repeated antibiotherapies lead to increasingly tolerant and hard-to-treat biofilms. In the context of growing antimicrobial resistance and restricted therapeutic options, antimicrobial photodynamic therapy (aPDT) shows great promise as an alternative to conventional antimicrobial modalities. Typically, aPDT consists in irradiating a non-toxic photosensitizer (PS) to generate reactive oxygen species (ROS), which kill pathogens in the surrounding environment. In a previous study, we reported that some ruthenium (II) complexes ([Ru(II)]) can mediate potent photodynamic inactivation (PDI) against planktonic cultures of Pseudomonas aeruginosa and Staphylococcus aureus clinical isolates. In the present work, [Ru(II)] were further assayed to evaluate their ability to photo-inactivate such bacteria under more complex experimental conditions better recapitulating the microenvironment in lung infected airways. Bacterial PDI was tentatively correlated with the properties of [Ru(II)] in biofilms, in mucus, and following diffusion across the latter. Altogether, the results obtained demonstrate the negative impacting role of mucus and biofilm components on [Ru(II)]-mediated PDT, following different possible mechanisms of action. Technical limitations were also identified that may be overcome, making this report a pilot for other similar studies. In conclusion, [Ru(II)] may be subjected to specific chemical engineering and/or drug formulation to adapt their properties to the harsh micro-environmental conditions of the infected respiratory tract.
Collapse
Affiliation(s)
- Raphaëlle Youf
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Rosy Ghanem
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, 29200, Brest, France
| | - Adeel Nasir
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Gilles Lemercier
- Université de Reims Champagne-Ardenne, UMR CNRS 7312, BP 1039, CEDEX 2, 51687, Reims, France
| | - Tristan Montier
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
- CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, 29200, Brest, France
- CHU de Brest, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - Tony Le Gall
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200, Brest, France
| |
Collapse
|
18
|
Kaur K, Müller M, Müller M, Schönherr H. Photodynamic Eradication of Pseudomonas aeruginosa with Ru-Photosensitizers Encapsulated in Enzyme Degradable Nanocarriers. Pharmaceutics 2023; 15:2683. [PMID: 38140023 PMCID: PMC10747122 DOI: 10.3390/pharmaceutics15122683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
The development of new approaches for the treatment of the increasingly antibiotic-resistant pathogen Pseudomonas aeruginosa was targeted by enhancing the effect of local antimicrobial photodynamic therapy (aPDT) using poly(ethylene glycol)-block-poly(lactic acid) (PEG114-block-PLAx) nanocarriers that were loaded with a ruthenium-based photosensitizer (PS). The action of tris(1,10-phenanthroline) ruthenium (II) bis(hexafluorophosphate) (RuPhen3) encapsulated in PEG114-block-PLAx micelles and vesicles was shown to result in an appreciable aPDT inactivation efficiency against planktonic Pseudomonas aeruginosa. In particular, the encapsulation of the PS, its release, and the efficiency of singlet oxygen (1O2) generation upon irradiation with blue light were studied spectroscopically. The antimicrobial effect was analyzed with two strains of Pseudomonas aeruginosa. Compared with PS-loaded micelles, formulations of the PS-loaded vesicles showed 10 times enhanced activity with a strong photodynamic inactivation effect of at least a 4.7 log reduction against both a Pseudomonas aeruginosa lab strain and a clinical isolate collected from the lung of a cystic fibrosis (CF) patient. This work lays the foundation for the targeted eradication of Pseudomonas aeruginosa using aPDT in various medical application areas.
Collapse
Affiliation(s)
| | | | - Mareike Müller
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), Department of Chemistry and Biology, School of Science and Technology, University of Siegen, 57076 Siegen, Germany (M.M.)
| | - Holger Schönherr
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), Department of Chemistry and Biology, School of Science and Technology, University of Siegen, 57076 Siegen, Germany (M.M.)
| |
Collapse
|
19
|
Wojtala D, Kozieł S, Witwicki M, Niorettini A, Guz-Regner K, Bugla-Płoskońska G, Caramori S, Komarnicka UK. Antibactericidal Ir(III) and Ru(II) Complexes with Phosphine-Alkaloid Conjugate and Their Interactions with Biomolecules: A Case of N-Methylphenethylamine. Chemistry 2023; 29:e202301603. [PMID: 37584222 DOI: 10.1002/chem.202301603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/06/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
The phosphine ligand (Ph2 PCH2 N(CH3 )(CH2 )2 Ph, PNMPEA) obtained by the reaction of the (hydroxymethyl)diphenylphosphine with naturally occurring alkaloid N-methylphenethylamine, was used to synthesize the half-sandwich iridium(III) (Ir(η5 -Cp*)Cl2 Ph2 PCH2 N(CH3 )(CH2 )2 Ph, IrPNMPEA) and ruthenium(II) (Ru(η6 -p-cymene)Cl2 Ph2 PCH2 N(CH3 )(CH2 )2 Ph, RuPNMPEA) complexes. They were characterized using a vast array of methods, including 1D and 2D NMR, ESI(+)MS spectrometry, elemental analysis, cyclic voltammetry (CV), electron spectroscopy in the UV-Vis range (absorption, fluorescence) and density functional theory (DFT). The initial antimicrobial activity in vitro toward Gram-positive and Gram-negative bacterial strains was examined, indicating that both complexes are selective towards Gram-positive bacteria, e. g., Staphylococcus aureus, where the IrPNMPEA has been more bactericidal compared to RuPNMPEA. Additionally, the interactions of these compounds with various biomolecules, such as DNA (ctDNA, plasmid DNA, 9-ethylguanine (9-EtG), and 9-methyladenine (9-MeA)), nicotinamide adenine dinucleotide (NADH), glutathione (GSH), and ascorbic acid (Asc) were described. The results showed that both Ir(III) and Ru(II) complexes accelerate the oxidation process of NADH, GSH and Asc that appeared to occur by an electron transfer mechanism. Interestingly, only IrPNMPEA leads to the formation of various biomolecule adducts, which can explain its higher activity. Furthermore, RuPNMPEA and IrPNMPEA have been interacting with the DNA through weak noncovalent interactions.
Collapse
Affiliation(s)
- Daria Wojtala
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Sandra Kozieł
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Maciej Witwicki
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Alessandro Niorettini
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, University of Ferrara, Via L. Borsari 46, 44121, Ferrara, Italy
| | - Katarzyna Guz-Regner
- Department of Microbiology, Faculty of Biological Sciences, University of Wroclaw, Przybyszewskiego 63-77, 51-148, Wroclaw, Poland
| | - Gabriela Bugla-Płoskońska
- Department of Microbiology, Faculty of Biological Sciences, University of Wroclaw, Przybyszewskiego 63-77, 51-148, Wroclaw, Poland
| | - Stefano Caramori
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, University of Ferrara, Via L. Borsari 46, 44121, Ferrara, Italy
| | - Urszula K Komarnicka
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383, Wroclaw, Poland
| |
Collapse
|
20
|
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
|
21
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
22
|
Vorotnikov YA, Vorotnikova NA, Shestopalov MA. Silica-Based Materials Containing Inorganic Red/NIR Emitters and Their Application in Biomedicine. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5869. [PMID: 37687562 PMCID: PMC10488461 DOI: 10.3390/ma16175869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023]
Abstract
The low absorption of biological substances and living tissues in the red/near-infrared region (therapeutic window) makes luminophores emitting in the range of ~650-1350 nm favorable for in vitro and in vivo imaging. In contrast to commonly used organic dyes, inorganic red/NIR emitters, including ruthenium complexes, quantum dots, lanthanide compounds, and octahedral cluster complexes of molybdenum and tungsten, not only exhibit excellent emission in the desired region but also possess additional functional properties, such as photosensitization of the singlet oxygen generation process, upconversion luminescence, photoactivated effects, and so on. However, despite their outstanding functional applicability, they share the same drawback-instability in aqueous media under physiological conditions, especially without additional modifications. One of the most effective and thus widely used types of modification is incorporation into silica, which is (1) easy to obtain, (2) biocompatible, and (3) non-toxic. In addition, the variety of morphological characteristics, along with simple surface modification, provides room for creativity in the development of various multifunctional diagnostic/therapeutic platforms. In this review, we have highlighted biomedical applications of silica-based materials containing red/NIR-emitting compounds.
Collapse
Affiliation(s)
- Yuri A. Vorotnikov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev ave., 630090 Novosibirsk, Russia;
| | | | - Michael A. Shestopalov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Acad. Lavrentiev ave., 630090 Novosibirsk, Russia;
| |
Collapse
|
23
|
Labach DS, Kohio HP, Tse EA, Paparisto E, Friesen NJ, Pankovich J, Bazett M, Barr SD. The Metallodrug BOLD-100 Is a Potent Inhibitor of SARS-CoV-2 Replication and Has Broad-Acting Antiviral Activity. Biomolecules 2023; 13:1095. [PMID: 37509131 PMCID: PMC10377621 DOI: 10.3390/biom13071095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
The COVID-19 pandemic has highlighted an urgent need to discover and test new drugs to treat patients. Metal-based drugs are known to interact with DNA and/or a variety of proteins such as enzymes and transcription factors, some of which have been shown to exhibit anticancer and antimicrobial effects. BOLD-100 (sodium trans-[tetrachlorobis(1H-indazole)ruthenate(III)]dihydrate) is a novel ruthenium-based drug currently being evaluated in a Phase 1b/2a clinical trial for the treatment of advanced gastrointestinal cancer. Given that metal-based drugs are known to exhibit antimicrobial activities, we asked if BOLD-100 exhibits antiviral activity towards SARS-CoV-2. We demonstrated that BOLD-100 potently inhibits SARS-CoV-2 replication and cytopathic effects in vitro. An RNA sequencing analysis showed that BOLD-100 inhibits virus-induced transcriptional changes in infected cells. In addition, we showed that the antiviral activity of BOLD-100 is not specific for SARS-CoV-2, but also inhibits the replication of the evolutionarily divergent viruses Human Immunodeficiency Virus type 1 and Human Adenovirus type 5. This study identifies BOLD-100 as a potentially novel broad-acting antiviral drug.
Collapse
Affiliation(s)
- Daniel S Labach
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| | - Hinissan P Kohio
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| | - Edwin A Tse
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| | - Ermela Paparisto
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| | - Nicole J Friesen
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| | - Jim Pankovich
- Bold Therapeutics Inc., 422 Richards St, Suite 170, Vancouver, BC V6N 2Z4, Canada
| | - Mark Bazett
- Bold Therapeutics Inc., 422 Richards St, Suite 170, Vancouver, BC V6N 2Z4, Canada
| | - Stephen D Barr
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, Dental Sciences Building Room 3007, London, ON N6A 5C1, Canada
| |
Collapse
|
24
|
Marinescu G, Culita DC, Mocanu T, Mitran RA, Petrescu S, Stan MS, Chifiriuc MC, Popa M. New Nanostructured Materials Based on Mesoporous Silica Loaded with Ru(II)/Ru(III) Complexes with Anticancer and Antimicrobial Properties. Pharmaceutics 2023; 15:pharmaceutics15051458. [PMID: 37242698 DOI: 10.3390/pharmaceutics15051458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/08/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
A new series of nanostructured materials was obtained by functionalization of SBA-15 mesoporous silica with Ru(II) and Ru(III) complexes bearing Schiff base ligands derived from salicylaldehyde and various amines (1,2-diaminocyclohexane, 1,2-phenylenediamine, ethylenediamine, 1,3-diamino-2-propanol, N,N-dimethylethylenediamine, 2-aminomethyl-pyridine, and 2-(2-aminoethyl)-pyridine). The incorporation of ruthenium complexes into the porous structure of SBA-15 and the structural, morphological, and textural features of the resulting nanostructured materials were investigated by FTIR, XPS, TG/DTA, zeta potential, SEM, and N2 physisorption. The ruthenium complex-loaded SBA-15 silica samples were tested against A549 lung tumor cells and MRC-5 normal lung fibroblasts. A dose-dependent effect was observed, with the highest antitumoral efficiency being recorded for the material containing [Ru(Salen)(PPh3)Cl] (50%/90% decrease in the A549 cells' viability at a concentration of 70 μg/mL/200 μg/mL after 24 h incubation). The other hybrid materials have also shown good cytotoxicity against cancer cells, depending on the ligand included in the ruthenium complex. The antibacterial assay revealed an inhibitory effect for all samples, the most active being those containing [Ru(Salen)(PPh3)Cl], [Ru(Saldiam)(PPh3)Cl], and [Ru(Salaepy)(PPh3)Cl], especially against Staphylococcus aureus and Enterococcus faecalis Gram-positive strains. In conclusion, these nanostructured hybrid materials could represent valuable tools for the development of multi-pharmacologically active compounds with antiproliferative, antibacterial, and antibiofilm activity.
Collapse
Affiliation(s)
- Gabriela Marinescu
- Ilie Murgulescu Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Daniela C Culita
- Ilie Murgulescu Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Teodora Mocanu
- Ilie Murgulescu Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Raul-Augustin Mitran
- Ilie Murgulescu Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Simona Petrescu
- Ilie Murgulescu Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania
| | - Miruna S Stan
- Faculty of Biology, University of Bucharest, 90 Panduri Street, 050663 Bucharest, Romania
| | - Mariana C Chifiriuc
- Faculty of Biology, University of Bucharest, 90 Panduri Street, 050663 Bucharest, Romania
| | - Marcela Popa
- Faculty of Biology, University of Bucharest, 90 Panduri Street, 050663 Bucharest, Romania
| |
Collapse
|
25
|
Giacomazzo GE, Conti L, Fagorzi C, Pagliai M, Andreini C, Guerri A, Perito B, Mengoni A, Valtancoli B, Giorgi C. Ruthenium(II) Polypyridyl Complexes and Metronidazole Derivatives: A Powerful Combination in the Design of Photoresponsive Antibacterial Agents Effective under Hypoxic Conditions. Inorg Chem 2023; 62:7716-7727. [PMID: 37163381 DOI: 10.1021/acs.inorgchem.3c00214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Ruthenium(II) polypyridyl complexes (RPCs) are gaining momentum in photoactivated chemotherapy (PACT), thanks to the possibility of overcoming the classical reliance on molecular oxygen of photodynamic therapy while preserving the selective drug activation by using light. However, notwithstanding the intriguing perspectives, the translation of such an approach in the development of new antimicrobials has been only barely considered. Herein, MTZH-1 and MTZH-2, two novel analogues of metronidazole (MTZ), a mainstay drug in the treatment of anaerobic bacterial infections, were designed and inserted in the strained ruthenium complexes [Ru(tpy)(dmp)(MTZ-1)]PF6 (Ru2) and [Ru(tpy)(dmp)(MTZ-2)]PF6 (Ru3) (tpy = terpyridine, dmp = 2,9-dimethyl-1,10-phenanthroline) (Chart 1). Analogously to the parental compound [Ru(tpy)(dmp)(5NIM)]PF6 (Ru1) (5-nitroimidazolate), the Ru(II)-imidazolate coordination of MTZ derivatives resulted in promising Ru(II) photocages, capable to easily unleash the bioactive ligands upon light irradiation and increase the antibacterial activity against Bacillus subtilis, which was chosen as a model of Gram-positive bacteria. The photoreleased 5-nitroimidazole-based ligands led to remarkable phototoxicities under hypoxic conditions (<1% O2), with the lead compound Ru3 that exhibited the highest potency across the series, being comparable to the one of the clinical drug MTZ. Besides, the chemical architectures of MTZ derivatives made their interaction with NimAunfavorable, being NimA a model of reductases responsible for bacterial resistance against 5-nitroimidazole-based antibiotics, thus hinting at their possible use to combat antimicrobial resistance. This work may therefore provide fundamental knowledge in the design of novel photoresponsive tools to be used in the fight against infectious diseases. For the first time, the effectiveness of the "photorelease antimicrobial therapy" under therapeutically relevant hypoxic conditions was demonstrated.
Collapse
Affiliation(s)
- Gina Elena Giacomazzo
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Luca Conti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Camilla Fagorzi
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Marco Pagliai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Claudia Andreini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Annalisa Guerri
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Brunella Perito
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Alessio Mengoni
- Department of Biology, University of Florence, Via Madonna del Piano 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Barbara Valtancoli
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| | - Claudia Giorgi
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, Sesto Fiorentino, 50019 Firenze, Italy
| |
Collapse
|
26
|
Telli FC, Yavuz M, Denizaltı S, Salman Y. Study of Radiotherapy Properties and Antimicrobial Activity of Glyconanoparticles (GNPs) Generated from Imidazolium Salts. ChemistrySelect 2023. [DOI: 10.1002/slct.202203810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Affiliation(s)
- Fatma Cetin Telli
- Chemistry Department Ege University Faculty of Science 35100 Bornova Izmir Turkey
| | - Murat Yavuz
- Chemistry Department Dicle University Faculty of Science 21280 Sur Diyarbakir Turkey
| | - Serpil Denizaltı
- Chemistry Department Ege University Faculty of Science 35100 Bornova Izmir Turkey
| | - Yesim Salman
- Chemistry Department Ege University Faculty of Science 35100 Bornova Izmir Turkey
| |
Collapse
|
27
|
Ceramella J, Troiano R, Iacopetta D, Mariconda A, Pellegrino M, Catalano A, Saturnino C, Aquaro S, Sinicropi MS, Longo P. Synthesis of Novel N-Heterocyclic Carbene-Ruthenium (II) Complexes, “Precious” Tools with Antibacterial, Anticancer and Antioxidant Properties. Antibiotics (Basel) 2023; 12:antibiotics12040693. [PMID: 37107055 PMCID: PMC10135378 DOI: 10.3390/antibiotics12040693] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Ruthenium N-heterocyclic carbene (Ru-NHC) complexes show interesting physico-chemical properties as catalysts and potential in medicinal chemistry, exhibiting multiple biological activities, among them anticancer, antimicrobial, antioxidant, and anti-inflammatory. Herein, we designed and synthesized a new series of Ru-NHC complexes and evaluated their biological activities as anticancer, antibacterial, and antioxidant agents. Among the newly synthesized complexes, RANHC-V and RANHC-VI are the most active against triple-negative human breast cancer cell lines MDA-MB-231. These compounds were selective in vitro inhibitors of the human topoisomerase I activity and triggered cell death by apoptosis. Furthermore, the Ru-NHC complexes’ antimicrobial activity was studied against Gram-positive and -negative bacteria, revealing that all the complexes possessed the best antibacterial activity against the Gram-positive Staphylococcus aureus, at a concentration of 25 µg/mL. Finally, the antioxidant effect was assessed by DPPH and ABTS radicals scavenging assays, resulting in a higher ability for inhibiting the ABTS•+, with respect to the well-known antioxidant Trolox. Thus, this work provides encouraging insights for further development of novel Ru-NHC complexes as potent chemotherapeutic agents endowed with multiple biological properties.
Collapse
Affiliation(s)
- Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Rubina Troiano
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Stefano Aquaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
28
|
Singh S, Navale GR, Agrawal S, Singh HK, Singla L, Sarkar D, Sarma M, Choudhury AR, Ghosh K. Design and synthesis of ruthenium complexes and their studies on the inhibition of amyloid β (1-42) peptide aggregation. Int J Biol Macromol 2023; 239:124197. [PMID: 36972817 DOI: 10.1016/j.ijbiomac.2023.124197] [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: 11/10/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Misfolding and protein aggregation have been linked to numerous human neurodegenerative disorders such as Alzheimer's, prions, and Parkinson's. Due to their interesting photophysical properties, ruthenium (Ru) complexes have received considerable attention in studying protein aggregation. In this study, we synthesized the novel Ru complexes ([Ru(p-cymene)Cl(L-1)][PF6](Ru-1), and [Ru(p-cymene)Cl(L-2)][PF6](Ru-2)) and investigated their inhibitory activity against the bovine serum albumin (BSA) aggregation and the Aβ1-42 peptides amyloid formation. Several spectroscopic methods were used to characterize the complexes, and the molecular structure was determined by X-ray crystallography. Amyloid aggregation and inhibition activity were examined using the Thioflavin-T (ThT) assay, and secondary structures were analyzed by circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The cell viability assay was carried out on the neuroblastoma cell line, revealing that the Ru-2 complex showed better protective effects against Aβ1-42 peptide toxicity on neuro-2a cells than the Ru-1 complex. Molecular docking studies elucidate binding sites and interactions between the Ru-complexes and the Aβ1-42 fibrils. The experimental studies revealed that these complexes significantly inhibited BSA aggregation and Aβ1-42 amyloid fibril formation at 1:3 and 1:1 equimolar concentrations, respectively. Antioxidant assays demonstrated that these complexes act as antioxidants, protecting from amyloid-induced oxidative stress. Molecular docking studies with the monomeric Aβ1-42 (PDB: 1IYT) show hydrophobic interaction, and both complexes bind preferably in the central region of the peptide and coordinate with two binding sites of the peptide. Hence, we suggest that the Ru-based complexes could be applied as a potential agent in metallopharmaceutical research against Alzheimer's disease.
Collapse
Affiliation(s)
- Sain Singh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Govinda R Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Sonia Agrawal
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Haobam Kisan Singh
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Labhini Singla
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Dhiman Sarkar
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Manabendra Sarma
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Anghuman Roy Choudhury
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India; Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India.
| |
Collapse
|
29
|
Smitten K, Southam HM, Fairbanks S, Graf A, Chauvet A, Thomas JA. Clearing an ESKAPE Pathogen in a Model Organism; A Polypyridyl Ruthenium(II) Complex Theranostic that Treats a Resistant Acinetobacter baumannii Infection in Galleria mellonella. Chemistry 2023; 29:e202203555. [PMID: 36420820 PMCID: PMC10946903 DOI: 10.1002/chem.202203555] [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: 11/15/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022]
Abstract
In previous studies we have described the therapeutic action of luminescent dinuclear ruthenium(II) complexes based on the tetrapyridylphenazine, tpphz, bridging ligand on pathogenic strains of Escherichia coli and Enterococcus faecalis. Herein, the antimicrobial activity of the complex against pernicious Gram-negative ESKAPE pathogenic strains of Acinetobacter baumannii (AB12, AB16, AB184 and AB210) and Pseudomonas aeruginosa (PA2017, PA_ 007_ IMP and PA_ 004_ CRCN) are reported. Estimated minimum inhibitory concentrations and minimum bactericidal concentrations for the complexes revealed the complex shows potent activity against all A. baumannii strains, in both glucose defined minimal media and standard nutrient rich Mueller-Hinton-II. Although the activity was lower in P. aureginosa, a moderately high potency was observed and retained in carbapenem-resistant strains. Optical microscopy showed that the compound is rapidly internalized by A. baumannii. As previous reports had revealed the complex exhibited no toxicity in Galleria Mellonella up to concentrations of 80 mg/kg, the ability to clear pathogenic infection within this model was explored. The pathogenic concentrations to the larvae for each bacterium were determined to be≥105 for AB184 and≥103 CFU/mL for PA2017. It was found a single dose of the compound totally cleared a pathogenic A. baumannii infection from all treated G. mellonella within 96 h. Uniquely, in these conditions thanks to the imaging properties of the complex the clearance of the bacteria within the hemolymph of G. mellonella could be directly visualized through both optical and transmission electron microscopy.
Collapse
Affiliation(s)
- Kirsty Smitten
- Department of ChemistryUniversity of SheffieldSheffieldS3 7HFUK
| | | | - Simon Fairbanks
- Department of ChemistryUniversity of SheffieldSheffieldS3 7HFUK
| | - Arthur Graf
- Department of ChemistryUniversity of SheffieldSheffieldS3 7HFUK
| | - Adrien Chauvet
- Department of ChemistryUniversity of SheffieldSheffieldS3 7HFUK
| | - Jim A Thomas
- Department of ChemistryUniversity of SheffieldSheffieldS3 7HFUK
| |
Collapse
|
30
|
Catalano A, Mariconda A, Sinicropi MS, Ceramella J, Iacopetta D, Saturnino C, Longo P. Biological Activities of Ruthenium NHC Complexes: An Update. Antibiotics (Basel) 2023; 12:365. [PMID: 36830276 PMCID: PMC9952499 DOI: 10.3390/antibiotics12020365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Ruthenium N-heterocyclic carbene (NHC) complexes have unique physico-chemical properties as catalysts and a huge potential in medicinal chemistry and pharmacology, exhibiting a variety of notable biological activities. In this review, the most recent studies on ruthenium NHC complexes are summarized, focusing specifically on antimicrobial and antiproliferative activities. Ruthenium NHC complexes are generally active against Gram-positive bacteria, such as Bacillus subtilis, Staphylococcus aureus, Micrococcus luteus, Listeria monocytogenes and are seldom active against Gram-negative bacteria, including Salmonella typhimurium, Pseudomonas aeruginosa and Escherichia coli and fungal strains of Candida albicans. The antiproliferative activity was tested against cancer cell lines of human colon, breast, cervix, epidermis, liver and rat glioblastoma cell lines. Ruthenium NHC complexes generally demonstrated cytotoxicity higher than standard anticancer drugs. Further studies are needed to explore the mechanism of action of these interesting compounds.
Collapse
Affiliation(s)
- Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | | | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy
| |
Collapse
|
31
|
Jian Y, Peng Y, Zhou W, Xu Y, Li C, Wang X, Zhou Q. Ru(II) Complexes with Enaminone Structures for Rapid Sterilization of Staphylococcus aureus and MRSA with Little Accumulation of Drug Resistance. ChemMedChem 2023; 18:e202300065. [PMID: 36751034 DOI: 10.1002/cmdc.202300065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/09/2023]
Abstract
Drug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), pose a serious threat to human life. Therefore, there is urgent need to develop antibiotics with new chemical structures and antibacterial mechanisms, especially those that elicit little drug resistance after long-term use. Herein we synthesized three novel ruthenium complexes (Ru1-Ru3) containing the enaminone structures for the first time. At a concentration of 5 μM, Ru1-Ru3 can lead to a CFU reduction of about 5 log units towards S. aureus and MRSA. Interestingly, Ru3 displayed rapid bactericidal effects and could decrease the CFU numbers of both pathogens by 5 log units within 40 min. The control compounds (Ru4 and Ru5) without the enaminone structures displayed very poor antibacterial activity under the same conditions. Moreover, S. aureus did not show apparent drug resistance towards Ru3 after 20 passages incubation with a sublethal concentration. These results highlight the critical role of enaminone structures for antibacterial applications.
Collapse
Affiliation(s)
- Yao Jian
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yatong Peng
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Wanpeng Zhou
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yunli Xu
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Chao Li
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xuesong Wang
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Qianxiong Zhou
- Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| |
Collapse
|
32
|
Novel Organoruthenium(II) Complex C1 Selectively Inhibits Butyrylcholinesterase without Side Effects on Neuromuscular Transmission. Int J Mol Sci 2023; 24:ijms24032681. [PMID: 36769002 PMCID: PMC9916964 DOI: 10.3390/ijms24032681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Enzyme butyrylcholinesterase (BChE) shows increased activity in some brain regions after progression of Alzheimer's disease and is therefore one of the therapeutic targets for symptomatic treatment of this neurodegenerative disorder. The organoruthenium(II) complex [(η6-p-cymene)Ru(II)(1-hydroxy-3-methoxypyridine-2(1H)-thionato)pta]PF6 (C1) was designed based on the results of our previous structure-activity studies. Inhibitory activity toward cholinesterase enzymes shows that this complex selectively, competitively, and reversibly inhibits horse serum BChE (hsBChE) with an IC50 value of 2.88 µM. When tested at supra-pharmacological concentrations (30, 60, 90, and 120 µM), C1 had no significant effect on the maximal amplitude of nerve-evoked and directly elicited single-twitch and tetanic contractions. At the highest tested concentration (120 µM), C1 had no effect on resting membrane potential, but significantly decreased the amplitude of miniature end-plate potentials (MEPP) without reducing their frequency. The same concentration of C1 had no effect on the amplitude of end-plate potentials (EPP), however it shortened the half-decay time of MEPPs and EPPs. The decrease in the amplitude of MEPPs and shortening of the half-decay time of MEPPs and EPPs suggest a possible weak inhibitory effect on muscle-type nicotinic acetylcholine receptors (nAChR). These combined results show that, when applied at supra-pharmacological concentrations up to 120 µM, C1 does not importantly affect the physiology of neuromuscular transmission and skeletal muscle contraction.
Collapse
|
33
|
MacNeil GA, Chang SW, Prosser KE, Ye E, Heroux D, Lewis AR, Bally M, Walsby CJ. Spatial characterization of redox processes and speciation of Ru(III) anticancer complexes by 19F magnetic resonance imaging. Chem Commun (Camb) 2023; 59:623-626. [PMID: 36537324 DOI: 10.1039/d2cc04830b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The application of CF3-labeled Ru(III) anticancer complexes to magnetic resonance (MR) imaging of tumour tissues is demonstrated. By combining anatomical chemical-shift selective (CHESS) imaging with 19F chemical-shift imaging (CSI) MR methods, we show that oxidation states and ligand-exchange processes of the complexes can be spatially encoded. Measurements on different tissue models, including a human breast adenocarcinoma tumour, validate the application of these complexes as MR theranostics for the sensing and targeting of hypoxia.
Collapse
Affiliation(s)
- Gregory A MacNeil
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada.
| | - Stephanie W Chang
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada.
| | - Kathleen E Prosser
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada.
| | - Eric Ye
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada.
| | - Devon Heroux
- BC Cancer Research Institute, 675 West 10th Ave., Vancouver, BC, V5Z 1L3, Canada
| | - Andrew R Lewis
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada. .,Callaghan Innovation, 69 Gracefield Rd, Lower Hutt, 5010, New Zealand
| | - Marcel Bally
- BC Cancer Research Institute, 675 West 10th Ave., Vancouver, BC, V5Z 1L3, Canada
| | - Charles J Walsby
- Department of Chemistry, Simon Fraser University, 8888 University Dr., Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
34
|
Levina A, Chetcuti ARM, Lay PA. Controversial Role of Transferrin in the Transport of Ruthenium Anticancer Drugs. Biomolecules 2022; 12:biom12091319. [PMID: 36139158 PMCID: PMC9496346 DOI: 10.3390/biom12091319] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Ruthenium complexes are at the forefront of developments in metal-based anticancer drugs, but many questions remain open regarding their reactivity in biological media, including the role of transferrin (Tf) in their transport and cellular uptake. A well-known anticancer drug, KP1019 ((IndH)[RuIIICl4(Ind)2], where Ind = indazole) and a reference complex, [RuIII(nta)2]3- (nta = nitrilotriacetato(3-)) interacted differently with human apoTf, monoFeTf, or Fe2Tf. These reactions were studied by biolayer interferometry (BLI) measurements of Ru-Fe-Tf binding to recombinant human transferrin receptor 1 (TfR1) in conjunction with UV-vis spectroscopy and particle size analysis. Cellular Ru uptake in human hepatoma (HepG2) cells was measured under the conditions of the BLI assays. The mode of Tf binding and cellular Ru uptake were critically dependent on the nature of Ru complex, availability of Fe(III) binding sites of Tf, and the presence of proteins that competed for metal binding, particularly serum albumin. Cellular uptake of KP1019 was not Tf-mediated and occurred mostly by passive diffusion, which may also be suitable for treatments of inoperable cancers by intratumoral injections. High cellular Ru uptake from a combination of [RuIII(nta)2]3- and Fe2Tf in the absence of significant Ru-Tf binding was likely to be due to trapping of Ru(III) species into the endosome during TfR1-mediated endocytosis of Fe2Tf.
Collapse
Affiliation(s)
- Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: (A.L.); (P.A.L.)
| | | | - Peter A. Lay
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- School of Sydney Analytical, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: (A.L.); (P.A.L.)
| |
Collapse
|
35
|
Ma Y, Wei M, Wang X, Jiang L, Xiong Y, Cheng J, Tan Y, Liao X, Wang J. Synthesis and antibacterial against
S. aureus
of new ruthenium (II) polypyridine complexes containing pyrene groups. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yuanyuan Ma
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Ming Wei
- Kangda College of Nanjing Medical University Lianyungang Jiangsu China
| | - Xuerong Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Li Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Yanshi Xiong
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Jianxin Cheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences Guangxi Normal University Guilin People’s Republic of China
| | - Xiangwen Liao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| | - Jintao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation School of Pharmacy, Jiangxi Science&Technology Normal University Nanchang People’s Republic of China
| |
Collapse
|
36
|
Youf R, Nasir A, Müller M, Thétiot F, Haute T, Ghanem R, Jonas U, Schönherr H, Lemercier G, Montier T, Le Gall T. Ruthenium(II) Polypyridyl Complexes for Antimicrobial Photodynamic Therapy: Prospects for Application in Cystic Fibrosis Lung Airways. Pharmaceutics 2022; 14:pharmaceutics14081664. [PMID: 36015290 PMCID: PMC9412327 DOI: 10.3390/pharmaceutics14081664] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Antimicrobial photodynamic therapy (aPDT) depends on a variety of parameters notably related to the photosensitizers used, the pathogens to target and the environment to operate. In a previous study using a series of Ruthenium(II) polypyridyl ([Ru(II)]) complexes, we reported the importance of the chemical structure on both their photo-physical/physico-chemical properties and their efficacy for aPDT. By employing standard in vitro conditions, effective [Ru(II)]-mediated aPDT was demonstrated against planktonic cultures of Pseudomonas aeruginosa and Staphylococcus aureus strains notably isolated from the airways of Cystic Fibrosis (CF) patients. CF lung disease is characterized with many pathophysiological disorders that can compromise the effectiveness of antimicrobials. Taking this into account, the present study is an extension of our previous work, with the aim of further investigating [Ru(II)]-mediated aPDT under in vitro experimental settings approaching the conditions of infected airways in CF patients. Thus, we herein studied the isolated influence of a series of parameters (including increased osmotic strength, acidic pH, lower oxygen availability, artificial sputum medium and biofilm formation) on the properties of two selected [Ru(II)] complexes. Furthermore, these compounds were used to evaluate the possibility to photoinactivate P. aeruginosa while preserving an underlying epithelium of human bronchial epithelial cells. Altogether, our results provide substantial evidence for the relevance of [Ru(II)]-based aPDT in CF lung airways. Besides optimized nano-complexes, this study also highlights the various needs for translating such a challenging perspective into clinical practice.
Collapse
Affiliation(s)
- Raphaëlle Youf
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
| | - Adeel Nasir
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
| | - Mareike Müller
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), Department of Chemistry and Biology, University of Siegen, 57076 Siegen, Germany
| | - Franck Thétiot
- Unité Mixte de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 6521, Université de Brest (UBO), CS 93837, 29238 Brest, France
| | - Tanguy Haute
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
| | - Rosy Ghanem
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
| | - Ulrich Jonas
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, 57076 Siegen, Germany
| | - Holger Schönherr
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), Department of Chemistry and Biology, University of Siegen, 57076 Siegen, Germany
| | - Gilles Lemercier
- Coordination Chemistry Team, Unité Mixte de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7312, Institut de Chimie Moléculaire de Reims (ICMR), Université de Reims Champagne-Ardenne, BP 1039, CEDEX 2, 51687 Reims, France
| | - Tristan Montier
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
- CHRU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200 Brest, France
| | - Tony Le Gall
- INSERM, Univ Brest, EFS, UMR 1078, GGB-GTCA, 29200 Brest, France
- Correspondence:
| |
Collapse
|
37
|
Peña L, Jiménez C, Arancibia R, Angeli A, Supuran CT. Heterobimetallic complexes containing organometallic acylhydrazone ligands as potential inhibitors of human carbonic anhydrases. J Inorg Biochem 2022; 232:111814. [DOI: 10.1016/j.jinorgbio.2022.111814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022]
|
38
|
Flint KL, Huang DM, Linder-Patton OM, Sumby CJ, Keene FR. Synthesis of Triple‐Stranded Diruthenium(II) Compounds. Eur J Inorg Chem 2022. [DOI: 10.1002/ejic.202200225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Kate L. Flint
- The University of Adelaide Chemistry 5005 Adelaide AUSTRALIA
| | - David M. Huang
- The University of Adelaide Chemistry 5005 Adelaide AUSTRALIA
| | | | | | | |
Collapse
|
39
|
Levina A, Crans DC, Lay PA. Advantageous Reactivity of Unstable Metal Complexes: Potential Applications of Metal-Based Anticancer Drugs for Intratumoral Injections. Pharmaceutics 2022; 14:790. [PMID: 35456624 PMCID: PMC9026487 DOI: 10.3390/pharmaceutics14040790] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/30/2022] Open
Abstract
Injections of highly cytotoxic or immunomodulating drugs directly into the inoperable tumor is a procedure that is increasingly applied in the clinic and uses established Pt-based drugs. It is advantageous for less stable anticancer metal complexes that fail administration by the standard intravenous route. Such hydrophobic metal-containing complexes are rapidly taken up into cancer cells and cause cell death, while the release of their relatively non-toxic decomposition products into the blood has low systemic toxicity and, in some cases, may even be beneficial. This concept was recently proposed for V(V) complexes with hydrophobic organic ligands, but it can potentially be applied to other metal complexes, such as Ti(IV), Ga(III) and Ru(III) complexes, some of which were previously unsuccessful in human clinical trials when administered via intravenous injections. The potential beneficial effects include antidiabetic, neuroprotective and tissue-regenerating activities for V(V/IV); antimicrobial activities for Ga(III); and antimetastatic and potentially immunogenic activities for Ru(III). Utilizing organic ligands with limited stability under biological conditions, such as Schiff bases, further enhances the tuning of the reactivities of the metal complexes under the conditions of intratumoral injections. However, nanocarrier formulations are likely to be required for the delivery of unstable metal complexes into the tumor.
Collapse
Affiliation(s)
- Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Debbie C. Crans
- Department of Chemistry and the Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Peter A. Lay
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
40
|
Komarnicka UK, Niorettini A, Kozieł S, Pucelik B, Barzowska A, Wojtala D, Ziółkowska A, Lesiów M, Kyzioł A, Caramori S, Porchia M, Bieńko A. Two out of Three Musketeers Fight against Cancer: Synthesis, Physicochemical, and Biological Properties of Phosphino Cu I, Ru II, Ir III Complexes. Pharmaceuticals (Basel) 2022; 15:169. [PMID: 35215281 PMCID: PMC8876511 DOI: 10.3390/ph15020169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
Two novel phosphine ligands, Ph2PCH2N(CH2CH3)3 (1) and Ph2PCH2N(CH2CH2CH2CH3)2 (2), and six new metal (Cu(I), Ir(III) and Ru(II)) complexes with those ligands: iridium(III) complexes: Ir(η5-Cp*)Cl2(1) (1a), Ir(η5-Cp*)Cl2(2) (2a) (Cp*: Pentamethylcyclopentadienyl); ruthenium(II) complexes: Ru(η6-p-cymene)Cl2(1) (1b), Ru(η6-p-cymene)Cl2(2) (2b) and copper(I) complexes: [Cu(CH3CN)2(1)BF4] (1c), [Cu(CH3CN)2(2)BF4] (2c) were synthesized and characterized using elemental analysis, NMR spectroscopy, and ESI-MS spectrometry. Copper(I) complexes turned out to be highly unstable in the presence of atmospheric oxygen in contrast to ruthenium(II) and iridium(III) complexes. The studied Ru(II) and Ir(III) complexes exhibited promising cytotoxicity towards cancer cells in vitro with IC50 values significantly lower than that of the reference drug-cisplatin. Confocal microscopy analysis showed that Ru(II) and Ir(III) complexes effectively accumulate inside A549 cells with localization in cytoplasm and nuclei. A precise cytometric analysis provided clear evidence for the predominance of apoptosis in induced cell death. Furthermore, the complexes presumably induce the changes in the cell cycle leading to G2/M phase arrest in a dose-dependent manner. Gel electrophoresis experiments revealed that Ru(II) and Ir(III) inorganic compounds showed their unusual low genotoxicity towards plasmid DNA. Additionally, metal complexes were able to generate reactive oxygen species as a result of redox processes, proved by gel electrophoresis and cyclic voltamperometry. In vitro cytotoxicity assays were also carried out within multicellular tumor spheroids and efficient anticancer action on these 3D assemblies was demonstrated. It was proven that the hydrocarbon chain elongation of the phosphine ligand coordinated to the metal ions does not influence the cytotoxic effect of resulting complexes in contrast to metal ions type.
Collapse
Affiliation(s)
- Urszula K. Komarnicka
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| | - Alessandro Niorettini
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy; (A.N.); (S.C.)
| | - Sandra Kozieł
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| | - Barbara Pucelik
- Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland; (B.P.); (A.B.)
| | - Agata Barzowska
- Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387 Krakow, Poland; (B.P.); (A.B.)
| | - Daria Wojtala
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| | - Aleksandra Ziółkowska
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| | - Monika Lesiów
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| | - Agnieszka Kyzioł
- Faculty of Chemistry, Jagiellonian University in Krakow, Gronostajowa 2, 30-387 Krakow, Poland;
| | - Stefano Caramori
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy; (A.N.); (S.C.)
| | | | - Alina Bieńko
- Faculty of Chemistry, University of Wroclaw, Joliot-Curie 14, 50-383 Wroclaw, Poland; (S.K.); (D.W.); (A.Z.); (M.L.); (A.B.)
| |
Collapse
|
41
|
Potential applicability of Schiff bases and their metal complexes during COVID-19 pandemic – a review. REV INORG CHEM 2022. [DOI: 10.1515/revic-2021-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
The rapid growth and revolution in the area of emerging therapeutics has been able to save the life of millions of patients globally. Besides these developments, the microbes are consistently struggling for their own survival and hence becoming quite more sturdy and incurable to existing drugs. Covid-19 virus and Black Fungus are recent examples of failure of medical preparations and strength of these viruses beyond the imagination of medical practitioners. Henceforth the study has made an extensive survey of exiting literature on heterocyclic schiff bases and their transition metal complexes to look for their potential applicability as antimicrobial agents. The inherent physiognomies of the essential properties of these transition metal complexes including thermodynamic, kinetic and chelating are comparatively modifiable as per requirements. The study has found that the biological applications of these transition metal complexes are well suited to be used as antibacterial and antifungal agents.
Collapse
|
42
|
Navale G, Singh S, Agrawal S, Ghosh C, Roy Choudhury A, Roy P, Sarkar D, Ghosh K. DNA binding, antitubercular, antibacterial and anticancer studies of newly designed piano-stool ruthenium( ii) complexes. Dalton Trans 2022; 51:16371-16382. [DOI: 10.1039/d2dt02577a] [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
The chemotherapeutic potential of ruthenium(ii) complexes as DNA binding, antitubercular, antibacterial, and anticancer agents.
Collapse
Affiliation(s)
- Govinda Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Sain Singh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Sonia Agrawal
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Chandrachur Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India
| | - Angshuman Roy Choudhury
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Partha Roy
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India
| | - Dhiman Sarkar
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India
| |
Collapse
|
43
|
Stimulation of Sulfonamides Antibacterial Drugs Activity as a Result of Complexation with Ru(III): Physicochemical and Biological Study. Int J Mol Sci 2021; 22:ijms222413482. [PMID: 34948278 PMCID: PMC8708937 DOI: 10.3390/ijms222413482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/03/2021] [Accepted: 12/12/2021] [Indexed: 11/16/2022] Open
Abstract
Antibiotic resistance is a global problem, and one promising solution to overcome this issue is using metallodrugs, which are drugs containing metal ions and ligands. These complexes are superior to free ligands in various characteristics including anticancer properties and mechanism of action. The pharmacological potential of metallodrugs can be modulated by the appropriate selection of ligands and metal ions. A good example of proper coordination is the combination of sulfonamides (sulfamerazine, sulfathiazole) with a ruthenium(III) ion. This work aimed to confirm that the activity of sulfonamides antibacterial drugs is initiated and/or stimulated by their coordination to an Ru(III) ion. The study determined the structure, electrochemical profile, CT-DNA affinity, and antimicrobial as well as anticancer properties of the synthesized complexes. The results proved that Ru(III) complexes exhibited better biological properties than the free ligands.
Collapse
|
44
|
Youf R, Müller M, Balasini A, Thétiot F, Müller M, Hascoët A, Jonas U, Schönherr H, Lemercier G, Montier T, Le Gall T. Antimicrobial Photodynamic Therapy: Latest Developments with a Focus on Combinatory Strategies. Pharmaceutics 2021; 13:1995. [PMID: 34959277 PMCID: PMC8705969 DOI: 10.3390/pharmaceutics13121995] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial photodynamic therapy (aPDT) has become a fundamental tool in modern therapeutics, notably due to the expanding versatility of photosensitizers (PSs) and the numerous possibilities to combine aPDT with other antimicrobial treatments to combat localized infections. After revisiting the basic principles of aPDT, this review first highlights the current state of the art of curative or preventive aPDT applications with relevant clinical trials. In addition, the most recent developments in photochemistry and photophysics as well as advanced carrier systems in the context of aPDT are provided, with a focus on the latest generations of efficient and versatile PSs and the progress towards hybrid-multicomponent systems. In particular, deeper insight into combinatory aPDT approaches is afforded, involving non-radiative or other light-based modalities. Selected aPDT perspectives are outlined, pointing out new strategies to target and treat microorganisms. Finally, the review works out the evolution of the conceptually simple PDT methodology towards a much more sophisticated, integrated, and innovative technology as an important element of potent antimicrobial strategies.
Collapse
Affiliation(s)
- Raphaëlle Youf
- Univ Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France; (R.Y.); (A.H.); (T.M.)
| | - Max Müller
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology of Micro and Nanochemistry and Engineering (Cμ), Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Straße 2, 57076 Siegen, Germany; (M.M.); (M.M.)
| | - Ali Balasini
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Straße 2, 57076 Siegen, Germany; (A.B.); (U.J.)
| | - Franck Thétiot
- Unité Mixte de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 6521, Université de Brest (UBO), CS 93837, 29238 Brest, France
| | - Mareike Müller
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology of Micro and Nanochemistry and Engineering (Cμ), Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Straße 2, 57076 Siegen, Germany; (M.M.); (M.M.)
| | - Alizé Hascoët
- Univ Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France; (R.Y.); (A.H.); (T.M.)
| | - Ulrich Jonas
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Straße 2, 57076 Siegen, Germany; (A.B.); (U.J.)
| | - Holger Schönherr
- Physical Chemistry I & Research Center of Micro- and Nanochemistry and (Bio)Technology of Micro and Nanochemistry and Engineering (Cμ), Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Straße 2, 57076 Siegen, Germany; (M.M.); (M.M.)
| | - Gilles Lemercier
- Coordination Chemistry Team, Unité Mixte de Recherche (UMR), Centre National de la Recherche Scientifique (CNRS) 7312, Institut de Chimie Moléculaire de Reims (ICMR), Université de Reims Champagne-Ardenne, BP 1039, CEDEX 2, 51687 Reims, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France; (R.Y.); (A.H.); (T.M.)
- CHRU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200 Brest, France
| | - Tony Le Gall
- Univ Brest, INSERM, EFS, UMR 1078, GGB-GTCA, F-29200 Brest, France; (R.Y.); (A.H.); (T.M.)
| |
Collapse
|
45
|
Mihajlović-Lalić LE, Poljarević J, Grgurić-Šipka S. Metal complexes with α-picolinic acid frameworks and their antitumor activity. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2021.120582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
46
|
Cheng J, Hu J. Recent Advances on Carbon Monoxide Releasing Molecules for Antibacterial Applications. ChemMedChem 2021; 16:3628-3634. [PMID: 34613654 DOI: 10.1002/cmdc.202100555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Indexed: 12/26/2022]
Abstract
Carbon monoxide (CO) has been known as an endogenous signaling molecule in addition to an air pollutant. It plays a critical role in many physiological and pathological processes. Therefore, CO has been recognized as a potent therapeutic agent for the treatment of numerous diseases such as cancers, rheumatoid arthritis, and so on. Instead of direct CO inhalation, two main categories of CO-releasing molecules (CORMs) (i. e., metal carbonyls and nonmetallic CO donors) have been developed to safely and locally deliver CO to target tissues. In this minireview, we summarize the recent achievements of CORMs on antibacterial applications. It appears that the antibacterial activity of CORMs is different from CO gas, which is tightly correlated to not only the types of CORMs applied but also the tested bacterial strains. In some circumstances, the antibacterial mechanisms are debated and need to be clarified. We hope more attention can be paid to this emerging field and new antibacterial agents with a low risk of drug resistance can be developed.
Collapse
Affiliation(s)
- Jian Cheng
- Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, 96 Jinzhai Road, Anhui 230026, Hefei, China
| | - Jinming Hu
- Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, 96 Jinzhai Road, Anhui 230026, Hefei, China
| |
Collapse
|
47
|
Singh A, Kaushik V, Chahal S, Goswami A, Nain S. Efficient Degradation of Methylene Blue Dye and Antibacterial Performance of Shape Controlled RuO
2
Nanocomposites. ChemistrySelect 2021. [DOI: 10.1002/slct.202102546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Amanvir Singh
- Department of Chemistry Deenbandhu Chhotu Ram University of Science and Technology Murthal 131039 Haryana India
| | - Vikas Kaushik
- Department of Biotechnology Deenbandhu Chhotu Ram University of Science and Technology Murthal 131039 Haryana India
| | - Surjeet Chahal
- Department of Physics Deenbandhu Chhotu Ram University of Science and Technology Murthal 131039 Haryana India
| | - Arkaja Goswami
- Department of Chemistry Shyamlal College University of Delhi 110032 New Delhi India
| | - Sonia Nain
- Department of Chemistry Deenbandhu Chhotu Ram University of Science and Technology Murthal 131039 Haryana India
| |
Collapse
|
48
|
Jabłońska-Wawrzycka A, Rogala P, Czerwonka G, Gałczyńska K, Drabik M, Dańczuk M. Ruthenium Complexes with 2-Pyridin-2-yl-1 H-benzimidazole as Potential Antimicrobial Agents: Correlation between Chemical Properties and Anti-Biofilm Effects. Int J Mol Sci 2021; 22:10113. [PMID: 34576276 PMCID: PMC8471145 DOI: 10.3390/ijms221810113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022] Open
Abstract
Antimicrobial resistance is a growing public health concern that requires urgent action. Biofilm-associated resistance to antimicrobials begins at the attachment phase and increases as the biofilms maturate. Hence, interrupting the initial binding process of bacteria to surfaces is essential to effectively prevent biofilm-associated problems. Herein, we have evaluated the antibacterial and anti-biofilm activities of three ruthenium complexes in different oxidation states with 2-pyridin-2-yl-1H-benzimidazole (L1 = 2,2'-PyBIm): [(η6-p-cymene)RuIIClL1]PF6 (Ru(II) complex), mer-[RuIIICl3(CH3CN)L1]·L1·3H2O (Ru(III) complex), (H2L1)2[RuIIICl4(CH3CN)2]2[RuIVCl4(CH3CN)2]·2Cl·6H2O (Ru(III/IV) complex). The biological activity of the compounds was screened against Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa strains. The results indicated that the anti-biofilm activity of the Ru complexes at concentration of 1 mM was better than that of the ligand alone against the P. aeruginosa PAO1. It means that ligand, in combination with ruthenium ion, shows a synergistic effect. The effect of the Ru complexes on cell surface properties was determined by the contact angle and zeta potential values. The electric and physical properties of the microbial surface are useful tools for the examined aggregation phenomenon and disruption of the adhesion. Considering that intermolecular interactions are important and largely define the functions of compounds, we examined interactions in the crystals of the Ru complexes using the Hirshfeld surface analysis.
Collapse
Affiliation(s)
| | - Patrycja Rogala
- Institute of Chemistry, Jan Kochanowski University of Kielce, 7 Uniwersytecka Str., 25-406 Kielce, Poland;
| | - Grzegorz Czerwonka
- Institute of Biology, Jan Kochanowski University of Kielce, 7 Uniwersytecka Str., 25-406 Kielce, Poland; (G.C.); (K.G.)
| | - Katarzyna Gałczyńska
- Institute of Biology, Jan Kochanowski University of Kielce, 7 Uniwersytecka Str., 25-406 Kielce, Poland; (G.C.); (K.G.)
| | - Marcin Drabik
- Institute of Physics, Jan Kochanowski University of Kielce, 7 Uniwersytecka Str., 25-406 Kielce, Poland;
| | - Magdalena Dańczuk
- Faculty of Environmental, Geomatic and Energy Engineering, Kielce University of Technology, 7 Tysiąclecia Państwa Polskiego Ave., 25-314 Kielce, Poland;
| |
Collapse
|