1
|
Wang YC, Qian C, Peng ZL, Hou XJ, Wang LL, Chao H, Ji LN. Dual topoisomerase I and II poisoning by chiral Ru(II) complexes containing 2-thiophenylimidazo[4,5-f][1,10]phenanthroline derivatives. J Inorg Biochem 2013; 130:15-27. [PMID: 24145066 DOI: 10.1016/j.jinorgbio.2013.09.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/25/2013] [Accepted: 09/25/2013] [Indexed: 02/02/2023]
Abstract
A series of chiral Ru(II) complexes bearing thiophene ligands were synthesized and characterized. Both Ru(II) complexes Δ/Λ-[Ru(bpy)2(pscl)](2+) (Δ/Λ-1) and Δ/Λ-[Ru(bpy)2(psbr)](2+) (Δ/Λ-2) (bpy=2,2'-bipyridine, pscl=2-(5-chlorothiophen-2-yl)imidazo[4,5-f][1,10]phenanthroline, psbr=2-(5-bromothiophen-2-yl)imidazo[4,5-f][1,10]phenanthroline) showed antitumor activities against A549, HepG2 and BEL-7402 tumor cell lines, especially HeLa tumor cell line. Moreover, Δ enantiomers were more active than Λ enantiomers, accounting for the different cellular uptake. In addition, with the extension of time, these enantiomers could finally accumulate in the nucleus, suggesting that nucleic acids were the cellular target of these enantiomers. The DNA-binding behaviors of complexes were studied using spectroscopic and viscosity measurements. Results suggested that four complexes could bind to DNA in an intercalative mode but no obvious DNA-binding selectivity between the enantiomers was observed. Topoisomerase inhibition and DNA religation assay confirmed that four complexes acted as efficient dual topoisomerase I and II poisons, DNA strand breaks had also been observed from alkaline single cell gel electrophoresis (comet assay). Δ-1 and Δ-2 inhibited the growth of HeLa cells through the induction of apoptotic cell death, as evidenced by the Alexa Fluor® 488 annexin V staining assays and flow cytometry analysis. The results demonstrated that Δ/Λ-1 and Δ/Λ-2 acted as dual topoisomerase I and II poisons and caused DNA damage that could lead to cell cycle arrest by apoptosis.
Collapse
|
Research Support, Non-U.S. Gov't |
12 |
44 |
2
|
Terpyridine and Quaterpyridine Complexes as Sensitizers for Photovoltaic Applications. MATERIALS 2016; 9:ma9030137. [PMID: 28773266 PMCID: PMC5456731 DOI: 10.3390/ma9030137] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/16/2016] [Accepted: 02/22/2016] [Indexed: 12/22/2022]
Abstract
Terpyridine and quaterpyridine-based complexes allow wide light harvesting of the solar spectrum. Terpyridines, with respect to bipyridines, allow for achieving metal-complexes with lower band gaps in the metal-to-ligand transition (MLCT), thus providing a better absorption at lower energy wavelengths resulting in an enhancement of the solar light-harvesting ability. Despite the wider absorption of the first tricarboxylate terpyridyl ligand-based complex, Black Dye (BD), dye-sensitized solar cell (DSC) performances are lower if compared with N719 or other optimized bipyridine-based complexes. To further improve BD performances several modifications have been carried out in recent years affecting each component of the complexes: terpyridines have been replaced by quaterpyridines; other metals were used instead of ruthenium, and thiocyanates have been replaced by different pinchers in order to achieve cyclometalated or heteroleptic complexes. The review provides a summary on design strategies, main synthetic routes, optical and photovoltaic properties of terpyridine and quaterpyridine ligands applied to photovoltaic, and focuses on n-type DSCs.
Collapse
|
Review |
9 |
40 |
3
|
Corrêa RS, da Silva MM, Graminha AE, Meira CS, Santos JAFD, Moreira DRM, Soares MBP, Von Poelhsitz G, Castellano EE, Bloch C, Cominetti MR, Batista AA. Ruthenium(II) complexes of 1,3-thiazolidine-2-thione: Cytotoxicity against tumor cells and anti-Trypanosoma cruzi activity enhanced upon combination with benznidazole. J Inorg Biochem 2016; 156:153-63. [PMID: 26795676 DOI: 10.1016/j.jinorgbio.2015.12.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 12/16/2015] [Accepted: 12/28/2015] [Indexed: 10/22/2022]
Abstract
Three new mixed and mononuclear Ru(II) complexes containing 1,3-thiazolidine-2-thione (tzdtH) were synthesized and characterized by spectroscopic analysis, molar conductivity, cyclic voltammetry, high-resolution electrospray ionization mass spectra and X-ray diffraction. The complexes presented unique stereochemistry and the proposed formulae are: [Ru(tzdt)(bipy)(dppb)]PF6 (1), cis-[Ru(tzdt)2(PPh3)2] (2) and trans-[Ru(tzdt)(PPh3)2(bipy)]PF6 (3), where dppb=1,4-bis(diphenylphosphino)butane and bipy=2,2'-bipyridine. These complexes demonstrated strong cytotoxicity against cancer cell lines when compared to cisplatin. Specifically, complex 2 was the most potent cytotoxic agent against MCF-7 breast cells, while complexes 1 and 3 were more active in DU-145 prostate cells. Binding of complexes to ctDNA was determined by UV-vis titration and viscosity measurements and revealed binding constant (Kb) values in range of 1.0-4.9×10(3)M(-1), which are characteristic of compounds possessing weak affinity to ctDNA. In addition, these complexes presented antiparasitic activity against Trypanosoma cruzi. Specifically, complex 3 demonstrated strong potency, moderate selectivity index and acted in synergism with the approved antiparasitic drug, benznidazole. Additionally, complex 3 caused parasite cell death through a necrotic process. In conclusion, we demonstrated that Ru(II) complexes have powerful pharmacological activity, while the metal-free tzdtH does not provoke the same outcome.
Collapse
|
Research Support, Non-U.S. Gov't |
9 |
37 |
4
|
Wang P, Dong R, Guo S, Zhao J, Zhang ZM, Lu TB. Improving photosensitization for photochemical CO 2-to-CO conversion. Natl Sci Rev 2020; 7:1459-1467. [PMID: 34691542 PMCID: PMC8288749 DOI: 10.1093/nsr/nwaa112] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/05/2019] [Accepted: 05/26/2020] [Indexed: 01/05/2023] Open
Abstract
Inspired by nature, improving photosensitization represents a vital direction for the development of artificial photosynthesis. The sensitization ability of photosensitizers (PSs) reflects in their electron-transfer ability, which highly depends on their excited-state lifetime and redox potential. Herein, for the first time, we put forward a facile strategy to improve sensitizing ability via finely tuning the excited state of Ru(II)-PSs (Ru-1–Ru-4) for efficient CO2 reduction. Remarkably, [Ru(Phen)2(3-pyrenylPhen)]2+ (Ru-3) exhibits the best sensitizing ability among Ru-1–Ru-4, over 17 times higher than that of typical Ru(Phen)32+. It can efficiently sensitize a dinuclear cobalt catalyst for CO2-to-CO conversion with a maximum turnover number of 66 480. Systematic investigations demonstrate that its long-lived excited state and suitable redox driving force greatly contributed to this superior sensitizing ability. This work provides a new insight into dramatically boosting photocatalytic CO2 reduction via improving photosensitization.
Collapse
|
|
5 |
25 |
5
|
Xue JJ, Bigdeli F, Liu JP, Hu ML, Morsali A. Ultrasonic-assisted synthesis and DNA interaction studies of two new Ru complexes; RuO 2 nanoparticles preparation. Nanomedicine (Lond) 2018; 13:2691-2708. [PMID: 30398084 DOI: 10.2217/nnm-2018-0174] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM To study of the interactions of two new ruthenium(II) complexes (C1 and C2) with calf thymus (CT)-DNA; production of RuO2 nanoparticles using the complexes precursor. MATERIALS & METHODS Complex C1 was characterized by x-ray crystallography. The binding of the complexes with (CT)-DNA was studied using techniques that include electronic absorption spectra, fluorescence and redox behavior. The preparation of RuO2 nanoparticles was carried out by thermal decomposition. RESULTS The interaction mode of DNA with complexes is the type of electrostatic. It was revealed that sonication of the samples, before thermal decomposition, has been affected the morphologies and sizes of the resulting nanoparticles. CONCLUSION The complexes are capable of interaction with DNA molecules and they have a good potential to prepare nanostructures.
Collapse
|
Research Support, Non-U.S. Gov't |
7 |
25 |
6
|
Chen Y, Bai L, Zhang P, Zhao H, Zhou Q. The Development of Ru(II)-Based Photoactivated Chemotherapy Agents. Molecules 2021; 26:5679. [PMID: 34577150 PMCID: PMC8465985 DOI: 10.3390/molecules26185679] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 01/10/2023] Open
Abstract
Photoactivated chemotherapy (PACT) is a novel cancer treatment method that has drawn increasing attention due to its high selectivity and low side effects by spatio-temporal control of irradiation. Compared with photodynamic therapy (PDT), oxygen-independent PACT is more suitable for treating hypoxic tumors. By finely tuning ligand structures and coordination configurations, many Ru(II) complexes can undergo photoinduced ligand dissociation, and the resulting Ru(II) aqua species and/or free ligands may have anticancer activity, showing their potential as PACT agents. In this mini-review, we summarized the progress in Ru(II)-based PACT agents, as well as challenges that researchers in this field still face.
Collapse
|
Review |
4 |
25 |
7
|
Anti-metastasis and anti-proliferation effect of mitochondria-accumulating ruthenium(II) complexes via redox homeostasis disturbance and energy depletion. J Inorg Biochem 2021; 217:111380. [PMID: 33578250 DOI: 10.1016/j.jinorgbio.2021.111380] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 01/10/2023]
Abstract
The antiproliferative activity of three cyclometalated Ru(II) complexes with the formula [Ru(bpy)2L]PF6, where bpy = 2,2'-bipyridine, Ru1: L1 = phenanthro[4,5-fgh]quinoxaline; Ru2: L2 = benzo[f]naphtho[2,1-h]quinoxaline; and Ru3: L3 = phenanthro[9,10-b]pyrazine, have been synthesized and characterized. The lipophilicity of the three Ru(II) complexes was modulated by the alteration of the planarity in the ligands of the complexes. With appropriate lipophilicity, Ru1-Ru3 exhibited mitochondrial accumulating property and cytotoxic activity against a spectrum of cancer cell lines. The underlying mechanism study indicated that these Ru(II) complexes can selectively accumulate in mitochondria and disrupt physiological processes, including the redox balance and energy generation in cancer cells. Elevation of iron content in triple-negative breast cancer (MDA-MB-231 cells) was observed after treatment with Ru(II) complexes, which may contribute to the production of reactive oxygen species (ROS) via Fenton reaction chemistry. Besides, the Ru(II) complexes decreased the intracellular glutathione (GSH) in cancer cells, leading to the failure in the cells to combat oxidative damage. Both of the mentioned processes contribute to the high oxidative stress and eventually lead to cancer cell death. On the other hand, Ru1-Ru3 significantly induced the depletion of adenosine triphosphate (ATP), causing disturbance of energy generation. Moreover, the results of wound-healing assay and transwell invasion assay, as well as the tube formation assay indicated the anti-migration and anti-angiogenesis properties of Ru1-Ru3. Our study demonstrated that these Ru(II) complexes are promising chemotherapeutic agents with oxidative stress induction and energy generation disturbance.
Collapse
|
Journal Article |
4 |
18 |
8
|
Wu M, Zhang Z, Yong J, Schenk PM, Tian D, Xu ZP, Zhang R. Determination and Imaging of Small Biomolecules and Ions Using Ruthenium(II) Complex-Based Chemosensors. Top Curr Chem (Cham) 2022; 380:29. [PMID: 35695976 PMCID: PMC9192387 DOI: 10.1007/s41061-022-00392-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 05/27/2022] [Indexed: 01/13/2023]
Abstract
Luminescence chemosensors are one of the most useful tools for the determination and imaging of small biomolecules and ions in situ in real time. Based on the unique photo-physical/-chemical properties of ruthenium(II) (Ru(II)) complexes, the development of Ru(II) complex-based chemosensors has attracted increasing attention in recent years, and thus many Ru(II) complexes have been designed and synthesized for the detection of ions and small biomolecules in biological and environmental samples. In this work, we summarize the research advances in the development of Ru(II) complex-based chemosensors for the determination of ions and small biomolecules, including anions, metal ions, reactive biomolecules and amino acids, with a particular focus on binding/reaction-based chemosensors for the investigation of intracellular analytes' evolution through luminescence analysis and imaging. The advances, challenges and future research directions in the development of Ru(II) complex-based chemosensors are also discussed.
Collapse
|
Review |
3 |
15 |
9
|
Eichhorn T, Kolbe F, Mišić S, Dimić D, Morgan I, Saoud M, Milenković D, Marković Z, Rüffer T, Dimitrić Marković J, Kaluđerović GN. Synthesis, Crystallographic Structure, Theoretical Analysis, Molecular Docking Studies, and Biological Activity Evaluation of Binuclear Ru(II)-1-Naphthylhydrazine Complex. Int J Mol Sci 2022; 24:ijms24010689. [PMID: 36614131 PMCID: PMC9821167 DOI: 10.3390/ijms24010689] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Ruthenium(II)-arene complexes have gained significant research interest due to their possible application in cancer therapy. In this contribution two new complexes are described, namely [{RuCl(η6-p-cymene)}2(μ-Cl)(μ-1-N,N'-naphthyl)]X (X = Cl, 1; PF6, 2), which were fully characterized by IR, NMR, and elemental microanalysis. Furthermore, the structure of 2 in the solid state was determined by a single crystal X-ray crystallographic study, confirming the composition of the crystals as 2·2MeOH. The Hirshfeld surface analysis was employed for the investigation of interactions that govern the crystal structure of 2·2MeOH. The structural data for 2 out of 2·2MeOH was used for the theoretical analysis of the cationic part [{RuCl(η6-p-cymene)}2(μ-Cl)(μ-1-N,N'-naphthyl)]+ (2a) which is common to both 1 and 2. The density functional theory, at B3LYP/6-31+G(d,p) basis set for H, C, N, and Cl atoms and LanL2DZ for Ru ions, was used for the optimization of the 2a structure. The natural bond orbital and quantum theory of atoms in molecules analyses were employed to quantify the intramolecular interactions. The reproduction of experimental IR and NMR spectra proved the applicability of the chosen level of theory. The binding of 1 to bovine serum albumin was examined by spectrofluorimetry and molecular docking, with complementary results obtained. Compound 1 acted as a radical scavenger towards DPPH• and HO• radicals, along with high activity towards cancer prostate and colon cell lines.
Collapse
|
research-article |
3 |
14 |
10
|
Li J, Sun Y, Xie L, He X, Tan L. Effect of ancillary ligands on the interaction of ruthenium(II) complexes with the triplex RNA poly(U)·poly(A)*poly(U). J Inorg Biochem 2014; 143:56-63. [PMID: 25528478 DOI: 10.1016/j.jinorgbio.2014.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 01/01/2023]
Abstract
Two new Ru(II) complexes with 1,8-naphthalimide group, [Ru(phen)2(pnip)](2+) (Ru1; phen=1,10-phenanthroline, pnip=2-[N-(p-phenyl)-1,8-napthalimide]imidazo[4',5'-f][1,10]phenanthroline) and [Ru(bpy)2(pnip)](2+) (Ru2; bpy=2,2'-bipyridine), have been synthesized and characterized. The interactions of Ru1 and Ru2 with the triplex RNA poly(U)•poly(A)*poly(U) (where • denotes the Watson-Crick base pairing and * denotes the Hoogsteen base pairing) were studied by various biophysical. Electronic spectra established that the binding affinity for Ru1 was greater than that for Ru2. Fluorescence and viscosity studies gave convincing evidence for a true intercalative binding of both complexes with the RNA triplex. UV melting studies confirmed that the two complexes could stabilize the triplex, whereas the effects of the two complexes on the stability of the Hoogsteen base-paired strand ploy(U) and the Watson-Crick base-paired duplex poly(U)•poly(A) of the triplex were different. In the case of Ru1, the increase of the thermal stability of the Hoogsteen base-paired strand was stronger than that of the Watson-Crick base-paired duplex. However, an opposite effect was observed in the case of Ru2. Circular dichroic studies suggested that the RNA triplex undergoes a conformational transition in the presence of Ru1, whereas the helicity of the RNA triplex still remains A-type in the presence of Ru2. The main results obtained here further advance our knowledge on the interaction of RNA triple-stranded structures with metal complexes, particularly ruthenium(II) complexes.
Collapse
|
Research Support, Non-U.S. Gov't |
11 |
14 |
11
|
Jin C, Bigdeli F, Liu KG, Ghasempour H, Hu ML, Morsali A. Sonochemical effect on two new Ruthenium(II) complexes with ligand (E)-N-((6-bromopyridin-2-yl)methylene)-4-(methylthio)aniline precursors for synthesis of RuO 2 nanoparticles. ULTRASONICS SONOCHEMISTRY 2017; 39:565-576. [PMID: 28732981 DOI: 10.1016/j.ultsonch.2017.05.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 06/07/2023]
Abstract
Two novel Ru(II) complexes [(η6-p-cymene)RuCl(L2)]PF6 (R2) and [(η6-C6H6)RuCl(L2)]PF6 (R4), with ligand (E)-N-((6-bromopyridin-2-yl)methylene)-4-(methylthio)aniline (L2), were synthesized and characterized by elemental analysis, 1H NMR, 13C NMR and IR spectroscopy. Based on X-ray crystallography studies, complexes R2 and R4 have coordination environments with formulated (η6-p-cymene)Ru(N2Cl) and (η6-C6H6)Ru(N2Cl), respectively. The thermal stabilities of compounds R2 and R4 were studied by thermal gravimetric (TG) and differential scanning calorimetry (DSC). Thermal decomposition of these complexes was at 280°C and 260°C under air atmosphere respectively. The interaction of these complexes with calf thymus DNA (CT-DNA) was explored through electronic absorption spectra, fluorescence and redox behavior studies. The results showed that the complexes bind to CT-DNA with electrostatic interactions. Nanoparticles of RuO2 were prepared by calcination of R2 and R4. Also the role of the ultrasound waves on the characteristics of the RuO2 nanoparticles was studied. The nanoparticles were characterized by IR spectroscopy and X-ray diffraction (XRD). Also size and morphology of nanoparticles were studied by scanning electron microscopy (SEM).
Collapse
|
|
8 |
12 |
12
|
Gano L, Pinheiro T, Matos AP, Tortosa F, Jorge TF, Gonçalves MS, Martins M, Morais TS, Valente A, Tomaz AI, Garcia MH, Marques F. Antitumour and Toxicity Evaluation of a Ru(II)-Cyclopentadienyl Complex in a Prostate Cancer Model by Imaging Tools. Anticancer Agents Med Chem 2020; 19:1262-1275. [PMID: 30887931 DOI: 10.2174/1871520619666190318152726] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/19/2018] [Accepted: 03/06/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND Ruthenium complexes have been extensively investigated for their prospective value as alternatives to cisplatin. Recently, we reported the in vitro anticancer properties of a family of organometallic ruthenium( II)-cyclopentadienyl complexes and have explored their mechanism of action. OBJECTIVE The purpose of this study was to evaluate the in vivo antitumour efficacy and toxicity of one of these Ru(II) compounds, [RuCp(mTPPMSNa)(2,2'-bipy)][CF3SO2] (TM85) which displayed an interesting spectrum of activity against several cancer cells. METHODS Studies to assess the antitumour activity and toxicity were performed in a metastatic prostate (PC3) mice model using ICP-MS, nuclear microscopy, elemental analysis and Transmission Electron Microscopy (TEM). RESULTS TM85 showed low systemic toxicity but no significant tumour reduction, when administered at tolerated dose (20mg/kg) over 10 days. Ru was mainly retained in the liver and less in kidneys, with low accumulation in tumour. Increased bilirubin levels, anomalous Ca and Fe concentrations in liver and mitochondria alterations were indicative of liver injury. The hepatotoxicity observed was less severe than that of cisplatin and no nephrotoxicity was found. CONCLUSION Under the experimental conditions of this study, TM85 is less toxic than cisplatin, induces similar tumour reduction and avoids the formation of metastatic foci. No renal toxicity was observed by the analysis of creatinine levels and the effective renal plasma flow by 99mTc-MAG3 clearance. Hence, it can be considered a valuable compound for further studies in the field of Ru-based anticancer drugs.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
12 |
13
|
Jin C, Bigdeli F, Jin ZM, Xie YR, Hu ML, Morsali A. Ultrasonic effect on RuO 2 nanostructures prepared by direct calcination of two new Ru(II)-organic supramolecular polymers. ULTRASONICS SONOCHEMISTRY 2017; 39:420-429. [PMID: 28732964 DOI: 10.1016/j.ultsonch.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 06/07/2023]
Abstract
Two novel Ru(II) complexes [(η6-p-cymene)RuCl(L1)]PF6 (R1) and [(η6-C6H6)RuCl(L1)]PF6 (R3) with ligand (E)-4-(methylthio)-N-((quinolin-2-yl)methylene)benzenamine (L1), were synthesized and investigated using elemental analysis, IR, 1H NMR, 13C NMR spectroscopy and X-ray crystallography. Complexes R1 and R3 have coordination environments with formulated (η6-p-cymene)Ru(N2Cl) and (η6-C6H6)Ru(N2Cl) respectively. The thermal stabilities of compounds R1 and R3 were studied by thermal gravimetric (TG) and differential scanning calorimetry (DSC). The binding of the complexes R1 and R3 to calf thymus DNA (CT DNA) was investigated by using electronic absorption spectra, fluorescence and redox behavior studies. Such experimental data showed that there are interactions between complexes and CT-DNA and the nature of this binding is electrostatic interaction type. Calcination of ultrasonic treated R1 and R3 results in the formation of nanoparticles of RuO2. The nanoparticles were characterized by IR spectroscopy and X-ray diffraction (XRD). Also size and morphology of nanoparticles were investigated by scanning electron microscopy (SEM).
Collapse
|
|
8 |
11 |
14
|
Feng Y, Liu X, Ma S, Wang F, Tan L. Ruthenium(II) polypyridyl complex [Ru(phen) 2dppz-idzo] 2+ as a colorimetric molecular "light switch" and powerful stabilizer for the RNA triplex poly(U)·poly(A)*poly(U). SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 212:240-245. [PMID: 30641364 DOI: 10.1016/j.saa.2018.12.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/09/2018] [Accepted: 12/27/2018] [Indexed: 06/09/2023]
Abstract
The interaction of [Ru(phen)2dppz-idzo]2+ (phen = 1,10-phenanthroline, dppz-idzo = dppz-imidazolone) with triplex RNA poly(U)·poly(A)*poly(U) was carried out by using spectroscopic and viscometric techniques in this work. Luminescent titrations suggest that [Ru(phen)2dppz-idzo]2+ shows better selectivity for poly(U)·poly(A)*poly(U) compared with poly(U)·poly(A) and poly(U), this complex exhibits a "light switch" effect with an emission enhancement factor of about 123 in the presence of poly(U)·poly(A)*poly(U). Significantly, this "light switch" behavior could even be observed by the naked eye under irradiation with UV light. To our knowledge, [Ru(bpy)2dppz-idzo]2+ is the first small molecule able to serve as a colorimetric molecular "light switch" for the triplex poly(U)·poly(A)*poly(U). Combined with the spectral and viscometric results as well as [Ru(phen)2dppz-idzo]2+ stabilizing the template duplex poly(U)·poly(A), we speculate that [Ru(phen)2dppz-idzo]2+ prefers to bind with the Hoogsteen base-paired strand (the third strand) of the triplex, thus the intercalating [Ru(phen)2dppz-idzo]2+ stabilizing the third strand is more marked in comparison with the Watson-Crick base-paired duplex of the triplex. The results obtained here may be useful for understanding the interaction of triplex RNA poly(U)·poly(A)*poly(U) with small molecule, particularly ruthenium(II) complexes.
Collapse
|
|
6 |
10 |
15
|
Pires WC, Lima BAV, de Castro Pereira F, Lima AP, Mello-Andrade F, Silva HD, da Silva MM, Colina-Vegas L, Ellena J, Batista AA, de Paul Silveira-Lacerda E. Ru(II)/diphenylphosphine/pyridine-6-thiolate complexes induce S-180 cell apoptosis through intrinsic mitochondrial pathway involving inhibition of Bcl-2 and p53/Bax activation. Mol Cell Biochem 2017; 438:199-217. [PMID: 28795366 DOI: 10.1007/s11010-017-3129-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/26/2017] [Indexed: 01/06/2023]
Abstract
The aim of this work was the synthesis, characterization, and cytotoxicity evaluation of three new Ru(II) complexes with a general formula [Ru(Spy)(bipy)(P-P)]PF6 [Spy = pyridine-6-thiolate; bipy = 2,2'-bipyridine; P-P = 1,2-bis(diphenylphosphine)ethane (1); 1,3-bis(diphenylphosphine) propane (2); and 1,1'-bis(diphenylphosphino)ferrocene] (4). Complex (3) with the 1,4-bis(diphenylphosphine)butane ligand, already known from the literature, was also synthesized, to be better studied here. The cytotoxicities of the complexes toward two kinds of cancerous cells (K562 and S-180 cells) were evaluated and compared to normal cells (L-929 and PBMC) by MTT assay. The complex [Ru(Spy)(bipy)(dppb)]PF6 (3) was selected to study both the cellular and molecular mechanisms underlying its promising anticancer action in S-180 cells. The results obtained from this study indicated that complex (3) induces cell cycle arrest in the G0/G1 phase in S-180 cells associated with a decrease in the number of cells in S phase. After 24 and 48 h of exposure to complex (3), the cell viability decreased when compared to the negative control. Complex (3) does not appear to be involved in the DNA damage, but induced changes in the mitochondrial membrane potential in S-180 cells. Furthermore, there was also an increase in the gene expression of Bax, Caspase 9, and Tp53. According to our results, complex (3) induces cell apoptosis through p53/Bax-dependent intrinsic pathway and suppresses the expression of active antiapoptotic Bcl-2 protein.
Collapse
|
Journal Article |
8 |
9 |
16
|
Liu SH, Zhu JW, Xu HH, Wang Y, Liu YM, Liang JB, Zhang GQ, Cao DH, Lin YY, Wu Y, Guo QF. Protein-binding, cytotoxicity in vitro and cell cycle arrest of ruthenium(II) polypyridyl complexes. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2016; 161:77-82. [PMID: 26956530 DOI: 10.1016/j.saa.2016.02.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 01/29/2016] [Accepted: 02/28/2016] [Indexed: 06/05/2023]
Abstract
The cytotoxic activity of two Ru(II) complexes against A549, BEL-7402, HeLa, PC-12, SGC-7901 and SiHa cell lines was investigated by MTT method. Complexes 1 and 2 show moderate cytotoxicity toward BEL-7402 cells with an IC50 value of 53.9 ± 3.4 and 39.3 ± 2.1 μM. The effects of the complexes inducing apoptosis, cellular uptake, reactive oxygen species and mitochondrial membrane potential in BEL-7402 cells have been studied by fluorescence microscopy. The percentages of apoptotic and necrotic cells and cell cycle arrest were studied by flow cytometry. The BSA-binding behaviors were investigated by UV/visible and fluorescent spectra.
Collapse
|
|
9 |
6 |
17
|
Hu H, Zhang H, Zhong R, Yang Y, Huang C, Chen J, Liang L, Chen Y, Liu Y. Synthesis, RNA-sequence and evaluation of anticancer efficacy of ruthenium(II) polypyridyl complexes toward HepG2 cells. J Inorg Biochem 2023; 244:112230. [PMID: 37084581 DOI: 10.1016/j.jinorgbio.2023.112230] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
In this article, four new Ru(II) complexes [Ru(dmbpy)2(TFBIP)](PF6)2 (dmbpy = 4,4'-dimethyl-2,2'-bipyridine, TFPIP = 2-(4'-trifluoromethyl)-[1,1'-biphenyl]-4-yl)-1H-imidazo[4,5-f][1,10]phenanthroline) (Ru1), [Ru(bpy)2(TFBIP)](PF6)2 (bpy = 2,2'-bipyridine) (Ru2), [Ru(phen)2(TFBIP)](PF6)2 (phen = 1,10-phenanthroline) (Ru3) and [Ru(dmp)2(TFBIP)](PF6)2 (dmp = 2,9-dimethyl-1,10-phenanthroline) (Ru4) were synthesized and characterized by elemental analysis, HRMS, IR, 1H NMR, 13C NMR and 19F NMR. The in vitro anticancer effect of the complexes on HepG2, A549, B16, HeLa, BEL-7402 and non-cancer LO2 cells was screened using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. The results illustrate that the complexes display moderate anticancer activity. Apoptotic assay with Annexin V/PI double staining method indicated that complexes induce apoptosis in HepG2 cells. Also, the complexes interfere with the mitochondrial functions, accompanied by the production of intracellular ROS as well as a reduction of mitochondrial membrane potential. The results obtained from the western blot demonstrated that the complexes upregulate pro-apoptotic Bax and downregulate anti-apoptotic Bcl-2, which further activates caspase 3 and promotes the cleavage of PARP. RNA-sequence showed that the complexes upregulate the expression of 40 genes and downregulate 66 genes. Antitumour in vivo demonstrated that Ru1 inhibits the tumor growth with a high inhibitory rate of 51.19%. Taken together, these results revealed that complexes Ru1, Ru2, Ru3 and Ru4 induce cell death in HepG2 cells via autophagy and a ROS-mediated mitochondrial apoptotic pathway.
Collapse
|
|
2 |
5 |
18
|
Sangiliapillai R, Arumugam R, Eswaran R, Seenivasan R. Micellar effect on the photophysics of heteroleptic ruthenium(II)-phenanthrolinedisulfonato complexes. LUMINESCENCE 2015; 31:30-7. [PMID: 25900090 DOI: 10.1002/bio.2917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 03/05/2015] [Accepted: 03/05/2015] [Indexed: 11/06/2022]
Abstract
Luminescent heteroleptic ruthenium(II) complexes of type RuLn X(3-n) [L = 1,10-phenanthroline (phen), X = 4,7 diphenyl phenanthroline disulfonate, (dpsphen) n = 0,1,2,3] were synthesized and their photophysical properties investigated in homogeneous and cationic (CTAB), anionic (SDS) and nonionic (Triton X-100) micelles. The luminescent quantum yield and lifetime of the complexes were found to increase in the presence of micellar media and on the introduction of a disulfonate ligand into the coordination sphere. Both electrostatic and hydrophobic interactions play an important role in the micellar media. Thus, by changing the nature of the ligands and the medium, we were able to tune the photophysical properties of Ru(II) complexes.
Collapse
|
|
10 |
4 |
19
|
Zhu X, Sun Q, Guo X, Liang C, Zhang Y, Huang W, Pei W, Huang Z, Chen L, Chen J. Cyclometalated ruthenium (II) complexes induced HeLa cell apoptosis through intracellular reductive injury. J Inorg Biochem 2023; 247:112333. [PMID: 37480763 DOI: 10.1016/j.jinorgbio.2023.112333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/29/2023] [Accepted: 07/14/2023] [Indexed: 07/24/2023]
Abstract
The main challenge of cancer chemotherapy is the resistance of tumor cells to oxidative damage. Herein, we proposed a novel antitumor strategy: cyclic metal‑ruthenium (Ru) complexes mediate reductive damage to kill tumor cells. We designed and synthesized Ru(II) complexes with β-carboline as ligands: [Ru (phen)2(NO2-Ph-βC)](PF6) (RuβC-7) and [Ru(phen)2(1-Ph-βC)](PF6) (RuβC-8). In vitro experimental results showed that RuβC-7 and RuβC-8 can inhibit cell proliferation, promote mitochondrial abnormalities, and induce DNA damage. Interestingly, RuβC-7 with SOD activity could reduce intracellular reactive oxygen species (ROS) levels, while RuβC-8 has the opposite effect. Accordingly, this study identified the reductive damage mechanism of tumor apoptosis, and may provide a new ideas for the design of novel metal complexes.
Collapse
|
|
2 |
3 |
20
|
Huang M, Zhang Y, Gong Y, Liang Z, Chen X, Ni Y, Pan X, Wu W, Chen J, Huang Z, Sun J. 8-Hydroxyquinoline ruthenium(II) complexes induce ferroptosis in HeLa cells by down-regulating GPX4 and ferritin. J Inorg Biochem 2023; 248:112365. [PMID: 37690267 DOI: 10.1016/j.jinorgbio.2023.112365] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Ruthenium complexes are one of the most promising anticancer drugs triggered extensive research. Here, the synthesis and characterization of two ruthenium(II) polypyridine complexes containing 8-hydroxylquinoline as ligand, [Ru(dip)2(8HQ)]PF6 (Ru1), [Ru(dpq)2(8HQ)]PF6 (Ru2) (8HQ = 8-hydroxylquinoline; dip = 4,7-diphenyl-1,10-phenanthroline; dpq = pyrazino[2,3-f][1,10]phenanthroline) were reported. On the basis of cytotoxicity tests, Ru1 (IC50 = 1.98 ± 0.02 μM) and Ru2 (IC50 = 10.02 ± 0.19 μM) both showed good anticancer activity in a panel of cell lines, especially in HeLa cells. Researches on mechanism indicated that Ru1 and Ru2 acted on mitochondria and nuclei and induced reactive oxygen species (ROS) accumulation, while the morphology of nuclei and cell cycle had no significant change. Western blot assay further proved that GPX4 and Ferritin were down-regulated, which eventually triggered ferroptosis in HeLa cells. In addition, the toxicity test of zebrafish embryos showed that the concentrations of Ru1 and Ru2 below 120 μM and 60 μM were safe and did not have obvious effect on the normal development of zebrafish embryos.
Collapse
|
|
2 |
2 |
21
|
Huang C, Zhang H, Yang Y, Liu H, Chen J, Wang Y, Liang L, Hu H, Liu Y. Synthesis, characterization, molecular docking, RNA-sequence and anticancer efficacy evaluation in vitro of ruthenium(II) complexes on B16 cells. J Inorg Biochem 2023; 247:112329. [PMID: 37478780 DOI: 10.1016/j.jinorgbio.2023.112329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
In recent years, the studies of the ruthenium(II) complexes on anticancer activity have been paid great attention, many Ru(II) complexes possess high anticancer efficiency. In this paper, three ligands CPIP (2-(4-chlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline), DCPIP (2-(3,4-dichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline), TCPIP (2-(2,3,5-trichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) and their three ruthenium (II) complexes [Ru(dip)2(CPIP)](PF6)2 (1, dip = 4,7-diphenyl-1,10-phenanthroline), [Ru(dip)2(DCPIP)](PF6)2 (2) and [Ru(dip)2(TCPIP)](PF6)2 (3) were synthesized and characterized. 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) assay was used to investigate in vitro cytotoxicity of complexes against various cancer cells. The results showed that complexes 1-3 exhibited pronounced cytotoxic effect on B16 cells with low IC50 values of 7.2 ± 0.1, 11.7 ± 0.6 and 1.2 ± 0.2 μM, respectively. The 3D model demonstrated that the complexes can validly prevent the cell proliferation. Apoptosis determined using Annexin V-FITC/PI double staining revealed that complexes 1-3 can effectively induce apoptosis in B16 cells. The intracellular localization of 1-3 in the mitochondria, the levels of intracellular reactive oxygen species (ROS), the opening of mitochondrial permeability transition pore as well as the decline of mitochondrial membrane potential were investigated, which demonstrated that the complexes 1-3 led to apoptosis via a ROS-mediated mitochondrial dysfunction pathway. The RNA-sequence indicated that the complexes upregulate the expression of 74 genes and downregulate the expression of 81 genes. The molecular docking showed that the complexes interact with the proteins through hydrogen bond, π-cation and π-π interaction. The results show that ruthenium(II) complexes 1, 2 and 3 can block tumor cell growth and induce cell death through autophagy and ROS-mediated mitochondrial dysfunction pathways.
Collapse
|
|
2 |
1 |
22
|
Yuan F, Liu X, Tan L. Binding properties of ruthenium(II) complexes [Ru(phen) 2(7-R-dppz)] 2+ (R = methyl or bromine) toward poly(U)•poly(A) RNA duplex. Int J Biol Macromol 2022; 209:1648-1655. [PMID: 35489619 DOI: 10.1016/j.ijbiomac.2022.04.091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022]
Abstract
Two Ru(II) complexes containing different substituents, [Ru(phen)2(7-CH3-dppz)]2+ (Ru1) and [Ru(phen)2(7-Br-dppz)]2+ (Ru2), have been synthesized in this study. The binding properties of Ru1 and Ru2 with the duplex RNA poly(U)•poly(A) (where "•" denotes the Watson - Crick base pairing) have been researched by biophysical techniques and viscosity measurements. Analysis of spectral titrations and viscosity measurements indicate that Ru1 and Ru2 bind to the duplex via intercalative, and the binding affinity of Ru1 with the duplex is remarkably higher than that of Ru2. Furthermore, fluorescence emission spectra demonstrates that although complexes Ru1 and Ru2 can act as molecular "light switches" for the duplex RNA, alters in fluorescence emission of Ru1 and Ru2 are prominent differences, and the effectiveness of Ru1 is more remarkable compared with that of Ru2. The melting experiments suggest that the duplex RNA stabilizing effects of Ru1 and Ru2 differ from each other, among them, complex Ru1 can obviously enhance the stability of the duplex RNA, while Ru2 has only a slightly stabilizing effect for the duplex RNA, indicating that Ru1 preferentially binds to RNA duplex over Ru2. The obtained results indicate that subtle modifications of the intercalative ligand of Ru(II) polypyridyl complex with either methyl or bromide group have a significant effect on the duplex-binding discrimination.
Collapse
|
|
3 |
1 |
23
|
Pan N, Zhang Y, Huang M, Liang Z, Gong Y, Chen X, Li Y, Wu C, Huang Z, Sun J. Lysosome-targeted ruthenium(II) complex encapsulated with pluronic ® F-127 induces oncosis in A549 cells. J Biol Inorg Chem 2024; 29:265-278. [PMID: 38189962 DOI: 10.1007/s00775-023-02039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/09/2023] [Indexed: 01/09/2024]
Abstract
Transition metal complexes with characteristics of unique packaging in nanoparticles and remarkable cancer cell cytotoxicity have emerged as potential alternatives to platinum-based antitumor drugs. Here we report the synthesis, characterization, and antitumor activities of three new Ruthenium complexes that introduce 5-fluorouracil-derived ligands. Notably, encapsulation of one such metal complex, Ru3, within pluronic® F-127 micelles (Ru3-M) significantly enhanced Ru3 cytotoxicity toward A549 cells by a factor of four. To determine the mechanisms underlying Ru3-M cytotoxicity, additional in vitro experiments were conducted that revealed A549 cell treatment with lysosome-targeting Ru3-M triggered oxidative stress, induced mitochondrial membrane potential depolarization, and drastically reduced intracellular ATP levels. Taken together, these results demonstrated that Ru3-M killed cells mainly via a non-apoptotic pathway known as oncosis, as evidenced by observed Ru3-M-induced cellular morphological changes including cytosolic flushing, cell swelling, and cytoplasmic vacuolation. In turn, these changes together caused cytoskeletal collapse and activation of porimin and calpain1 proteins with known oncotic functions that distinguished this oncotic process from other cell death processes. In summary, Ru3-M is a potential anticancer agent that kills A549 cells via a novel mechanism involving Ru(II) complex triggering of cell death via oncosis.
Collapse
|
|
1 |
|
24
|
Kushwaha R, Banerjee S. Dinuclear Ru(II) Complexes Containing Tetrapyrido[3,2-a : 2,3-c : 3,2-h : 2''',3'''-j]Phenazine Ligand for Biomedical Applications. Chembiochem 2025; 26:e202400931. [PMID: 39663208 DOI: 10.1002/cbic.202400931] [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: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Ruthenium complexes are among the most extensively studied and developed luminescent transition-metal complexes for anticancer applications. Dinuclear Ru(II) complexes have caught significant interest for larger size, higher charge, and variable complex shapes. In this concept, we have explored past and recent works on the possible biological applications of versatile tetrapyrido[3,2-a : 2,3-c : 3,2-h : 2''',3'''-j]phenazine (tppz)-based dinuclear Ru(II) complexes with a focus on their use as quadruplex DNA probes, organelle imaging, and phototherapeutic agents. This concept also points out that a particular type of dinuclear Ru(II) complexes can act as multitargeting and multifunctional anticancer agents -making this an exciting research area in which an array of further applications will likely emerge.
Collapse
|
Review |
1 |
|
25
|
Xu Y, Da X, Jian Y, Zhou W, Wu A, Wu Y, Peng Y, Liu X, Shi Y, Wang X, Zhou Q. A highly positively charged Ru(II) complex with photo-labile ligands for selective and efficient photo-inactivation of intracellular Staphylococcus aureus. J Inorg Biochem 2025; 268:112908. [PMID: 40209460 DOI: 10.1016/j.jinorgbio.2025.112908] [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: 12/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Due to the protection afforded by host cells, intracellular Staphylococcus aureus (S. aureus), particularly methicillin-resistant S. aureus (MRSA), poses a significantly greater challenge to eliminate compared to the extracellular counterparts. It is highly desirable to develop novel antibacterial agents which are capable of selectively and efficiently eradicating intracellular bacteria, including drug-resistant strains, while being less prone to induce bacterial resistance. In this work, two Ru(II) complexes (Ru1 and Ru2) with photo-labile ligands were designed and synthesized. Both Ru1 and Ru2 could covalently bind to DNA after photo-induced ligand dissociation. Compared to Ru1, the incorporation of a triphenylamine group adorned with two positively charged cationic pyridine units significantly boosts the DNA binding constant, bacterial binding/uptake level, and subsequently, the antibacterial activity of Ru2. Ru2 could selectively photo-inactivate intracellular S. aureus and MRSA, being more efficient than vancomycin both in vitro and in vivo. Interestingly, after 20 days' treatment at sublethal concentrations, S. aureus cells exhibited no obvious drug resistance towards Ru2 upon irradiation. Such appealing results may provide new sights for developing novel antibacterial agents against intractable intracellular pathogens and also prevalent drug resistance.
Collapse
|
|
1 |
|