Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Dalton Trans ; 50(44): 16311-16325, 2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34730582

ABSTRACT

Six different acylthiourea ligands (L1-L6) and their corresponding Ru(II)-p-cymene complexes (P1-P6) were designed to explore the structure-activity relationship of the complexes upon aliphatic chain and aromatic conjugation on the C- and N-terminals, respectively. The compounds were synthesized and adequately characterized using various analytical and spectroscopic techniques. The structures of P2-P6, solved using single crystal X-ray diffraction (XRD), confirmed the neutral monodentate coordination of the S atoms of the acylthiourea ligands to Ru(II) ions. In silico studies showed an increase of lipophilicity for the ligands with an increase in alkyl chain length or aromatic conjugation at the C- or N-terminal, respectively. Subsequently, mitogen-activated protein kinases (MAPK) were predicted as one of the primary targets for the complexes, which showed good binding affinity towards extracellular signal-regulated kinases (ERK1, ERK2 and ERK5), c-Jun N-terminal kinase (JNK) and p38 of the MAPK pathway. Henceforth, the complexes were tested for their anticancer activity in lung carcinoma (A549) and cisplatin-resistant lung carcinoma (cisA549R) cells and human umbilical vein epithelial normal cells (HUVEC). Interestingly, an increase in chain length or aromatic conjugation led to an increase in the activity of the complexes, with P5 (7.73 and 13.04 µM) and P6 (6.52 and 14.45 µM) showing the highest activity in A549 and cisA549R cells, which is better than the positive control, cisplatin (8.72 and 44.28 µM). Remarkably, we report the highest activity yet observed for complexes of the type [(η6-p-cymene)RuIICl2(S-acylthiourea)] in the tested cell lines. Aqueous solution studies showed that complexes P5 and P6 are rapidly hydrolyzed to produce solely aquated species that remained stable for 24 h. Staining assays and flow cytometric analyses of P5 and P6 in A549 cells revealed that the complexes induced apoptosis and arrested the cell cycle predominantly in the S phase. In vivo studies demonstrated the higher toxicity of cisplatin and a comparatively higher survival rate of mice injected with the most active complex P6. Histological analyses revealed that treatment with P6 at high doses of up to 8 mg kg-1 did not cause any palpable damage to the tested organs.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Cymenes , Ruthenium , Thioamides , Thiourea , A549 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Biological Availability , Cell Survival/drug effects , Coordination Complexes/administration & dosage , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Cymenes/administration & dosage , Cymenes/chemistry , Cymenes/pharmacokinetics , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intestinal Absorption , Ligands , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism , Molecular Docking Simulation , Ruthenium/administration & dosage , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Thioamides/administration & dosage , Thioamides/chemistry , Thioamides/pharmacokinetics , Thiourea/administration & dosage , Thiourea/chemistry , Thiourea/pharmacokinetics
2.
J Inorg Biochem ; 208: 111104, 2020 07.
Article in English | MEDLINE | ID: mdl-32485635

ABSTRACT

Ruthenium complexes are expected to be new opportunities for the development of antitumor agents. Herein, four ruthenium polypyridyl complexes ([Ru(bpy)2(CAPIP)](ClO4)2 (Ru(II)-1, bpy = 2,2'-bipyridine; CAPIP = (E)-2-(2-(furan-2-yl)vinyl)-1H-imidazo[4,5-f][1,10]phenanthroline), [Ru(phen)2(CA-PIP)](ClO4)2 (Ru(II)-2, phen = 1,10-phenanthroline), [Ru(dmb)2(CAPIP)](ClO4)2 (Ru(II)-3, dmb = 4,4'-dimethyl-2,2'-bipyridine), [Ru(dmb)2(ETPIP)](ClO4)2 (Ru(II)-4, ETPIP = 2-(4-(thiophen-2-ylethynyl)phenyl)-1H-imidazo[4,5-f][1,10]phen-anthroline)) have been investigated as mitochondria-targeted antitumor metallodrugs. DNA binding studies indicated that target Ru(II) complexes interacts with CT DNA (calf thymus DNA) by an intercalative mode. Cytotoxicity assay results demonstrate that Ru(II) complexes show high cytotoxicity against A549 cells with low IC50 value of 23.6 ± 2.3, 20.1 ± 1.9, 22.7 ± 1.8 and 18.4 ± 2.3 µM, respectively. Flow cytometry and morphological analysis revealed that these Ru(II) complexes can induce apoptosis in A549 cells. Intracellular reactive oxygen species (ROS) and mitochondrial membrane potential were also investigated by ImageXpress Micro XLS system. The experimental results indicate that the reactive oxygen species in A549 cells increased significantly and mitochondrial membrane potential decreased obviously. In addition, colocalization studies shown these complexes could get to the cytoplasm through the cell membrane and accumulate in the mitochondria. Furthermore, Ru(II) complexes can effectively induces cell cycle arrest at the S phase in A549 cells. Finally, cell invasion assay and quantitative studies were also performed to investigate the mechanism of this process. All in together, this study suggested that these Ru(II) complexes could induce apoptosis in A549 cells through cell cycle arrest and ROS-mediated mitochondrial dysfunction pathway.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Neoplasms , Pyridines , Ruthenium , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology
3.
J Inorg Biochem ; 208: 111099, 2020 07.
Article in English | MEDLINE | ID: mdl-32460056

ABSTRACT

A series of ruthenium(II)-arene complexes of several bipyridine and phenanthroline derivatives have been synthesized by employing a green and efficient protocol involving water as a solvent under sonication. The structures of all the complexes were elucidated by the spectroscopic analysis. The geometry of the chlorido and PTA (1,3,5-Triaza-7-phosphaadamantane) complexes were further confirmed by DFT and single crystal XRD. The stability study in various solvents, specifically in the intracellular one was conducted. Most of the compounds exhibited significant potency and selectivity against MCF7 and HeLa cell lines with respect to normal HEK-293 cells compared to cisplatin and RAPTA-C (Ruthenium(II)-arene PTA complex). Complex [(η6-hexamethylbenzene)RuCl(κ2-N,N-4,4'-di-n-nonyl-2,2'-bpy)]Cl (3e) presented best anticancer profiles against all the human cancer cells. Interestingly, few complexes turned up to be highly fluorescent depicted by the quantum yield values. Remarkably, [(η6-p-cymene)RuCl(κ2-N,N-bpy)]Cl (3i) was identified as most significant anticancer theranostic agent interms of potency, selectivity and fluorescence quantum yield. This complex also represented itself as significant cellular imaging agent in live U-87 MG cells which was monitored by confocal microscope. Absorption and emission spectral studies of bypyridine and phenanthroline complex series revealed that the complexes interacted with calf thymus DNA through groove binding as well as intercalative mode. In addition to this, strong binding efficacy of these scaffolds wih BSA (Bovin Serum Albumin) also enhanced their transportation property inside the cells.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Luminescent Measurements , Neoplasms , Phenanthrolines , Pyridines , Ruthenium , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , DNA/chemistry , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phenanthrolines/chemistry , Phenanthrolines/pharmacokinetics , Phenanthrolines/pharmacology , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology
4.
Metallomics ; 12(4): 504-513, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32051986

ABSTRACT

Guanine-rich DNA sequences can spontaneously fold into four-stranded structures called G-quadruplexes (G4s). G4s have been identified extensively in the promoter regions of several proto-oncogenes, including c-myc, as well as telomeres. G4s have attracted an increasing amount of attention in the field of nanotechnology because of their use as versatile building blocks of DNA-based nanostructures. In this study, we report the self-assembly of c-myc G-quadruplex DNA controlled by a pair of chiral ruthenium(ii) complexes coordinated by 2-(4-phenyacetylenephenyl)-1H-imidazo[4,5f][1,10]phenanthroline (PBEPIP), Λ-[Ru(bpy)2(PBEPIP)](ClO4)2 (Λ-RM0627, bpy = bipyridine) and Δ-[Ru(bpy)2(PBEPIP)](ClO4)2 (Δ-RM0627). Λ-RM0627 could promote the high-order self-assembly of c-myc G-quadruplex DNA into a nanowire structure, whereas Δ-RM0627 could induce DNA condensation into G-quadruplex aggregates. Moreover, in vitro studies on human liver carcinoma HepG2 cells showed that the nanowire of c-myc G-quadruplex DNA promoted by Λ-RM0627 could be localized in the nuclei of cells, whereas the nanoparticle of c-myc G-quadruplex DNA generated by Δ-RM0627 was taken up and localized in the cytoplasm. This study provides examples of the enantioselective self-assembly of G4 DNA molecules controlled by chiral ruthenium(ii) complexes and suggests the potential applications of assembled nanostructures as non-viral DNA vectors for gene therapy.


Subject(s)
Cell Nucleus/metabolism , Coordination Complexes/metabolism , Cytoplasm/metabolism , Ruthenium/metabolism , Biological Transport , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , DNA/chemistry , DNA/metabolism , G-Quadruplexes , Hep G2 Cells , Humans , Nanoparticles/chemistry , Nanowires , Phenanthrolines/chemistry , Phenanthrolines/metabolism , Protein Binding , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/metabolism , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Stereoisomerism
5.
Nanoscale ; 11(39): 18209-18223, 2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31560010

ABSTRACT

Rheumatoid arthritis (RA) is a degenerative joint disease caused by autoimmunity; for the effective treatment of RA while avoiding the side effects of conventional drugs, we have proposed a new therapeutic strategy to eliminate the inflammatory response in RA by regulating the immune system that promotes the transformation of M1-type macrophages to M2-type macrophages. Herein, we designed and synthesized a core-shell nanocomposite (QRu-PLGA-RES-DS NPs), which showed an effective therapeutic effect on RA by accurately inducing the polarization of M2 macrophages. In this system, the quadrilateral ruthenium nanoparticles (QRuNPs) with a photothermal effect were utilized as a core and the thermosensitive molecular poly (lactic-co-glycolic acid) (PLGA) modified with the targeted molecule dextran sulfate (DS) was employed as a shell. Then, the nanocarrier QRu-PLGA-DS NPs effectively improved the water solubility and targeting of resveratrol (RES) through self-assembly. Therefore, the QRu-PLGA-RES-DS NPs significantly enhanced the ability of RES to reverse the M1 type macrophages to the M2 type macrophages through an accurate release. In vivo experiments further demonstrated that the QRu-PLGA-RES-DS NPs could effectively accumulate in the lesion area with an exogenous stimulus, and this significantly enhanced the transformation of the M2 type macrophages and decreased the recruitment of the M1 type macrophages. Furthermore, the QRu-PLGA-RES-DS NPs effectively treated RA by eliminating the inflammatory response; in addition, photoacoustic imaging (PA) of the QRu NPs provided image guidance for the distribution and analysis of nanomedicine in inflammatory tissues. Hence, this therapeutic strategy promotes the biological applications of Ru-based nanoparticles in disease treatment.


Subject(s)
Hyperthermia, Induced , Macrophages/metabolism , Nanocomposites , Phototherapy , Resveratrol , Rheumatic Fever/therapy , Animals , Human Umbilical Vein Endothelial Cells , Humans , Macrophages/pathology , Mice , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polyglycolic Acid/pharmacology , RAW 264.7 Cells , Resveratrol/pharmacokinetics , Resveratrol/pharmacology , Rheumatic Fever/metabolism , Rheumatic Fever/pathology , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology
6.
Acta Biomater ; 82: 143-157, 2018 12.
Article in English | MEDLINE | ID: mdl-30316026

ABSTRACT

The blood-brain barrier (BBB) and low targeting are major obstacles for the treatment of gliomas. Accordingly, overcoming the BBB and enhancing the targeting of drugs to the glioma area are key to achieving a good therapeutic effect. Here, we have developed the mesoporous ruthenium nanosystem RBT@MRN-SS-Tf/Apt with dual targeting function. Transferrin (Tf) and aptamer AS1411 (Apt) are grafted on the surfaces of mesoporous ruthenium nanoparticles (MRN) with high loading capacity. This is achieved via redox-cleavable disulfide bonds, serving as both a capping agent and a targeting ligand, enabling the effective penetration of the blood-brain barrier and targeting the glioma. In addition, RBT@MRN-SS-Tf/Apt can specifically kill glioma cells in vitro and in vivo. Moreover, anti-tumor drugs [Ru(bpy)2(tip)]2+ (RBT) will produce reactive oxygen species and induce apoptosis of tumor cells under laser irradiation, providing photodynamic therapy (PDT) for the treatment of gliomas, and further prolonging the median survival period. The study shows that this chemical photodynamic therapy nanosystem can be used as an efficient and powerful synergistic system for the treatment of brain tumors and other brain diseases of the central nervous system. STATEMENT OF SIGNIFICANCE: In order to overcome the blood-brain barrier and low targeting, and enhance the anti-glioma activities of nanodrugs. We have developed RBT@MRN-SS-Tf/Apt with dual targeting function. It is achieved release drug via redox-cleavable disulfide bonds, and enable the effective penetration of the blood-brain barrier and targeting the glioma. Moreover, anti-tumor drugs RBT will produce reactive oxygen species and induce apoptosis of tumor cells under laser irradiation, providing photodynamic therapy (PDT) for the treatment of gliomas, and further prolonging the median survival period. Therefore, this chemical photodynamic therapy nanosystem can be used as an efficient and powerful synergistic system for the treatment of brain tumors and other brain diseases of the central nervous system.


Subject(s)
Glioma , Nanoparticles , Photochemotherapy , Ruthenium , Transferrin , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology , Transferrin/chemistry , Transferrin/pharmacokinetics , Transferrin/pharmacology
7.
Metallomics ; 10(3): 388-396, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29513313

ABSTRACT

Laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) was used to study the spatial distribution of two metallodrugs with anticancer activities in vivo, namely the organoruthenium plecstatin-1 (1) and its isosteric osmium analogue (2), in liver, kidneys, muscles and tumours of treated mice bearing a CT-26 tumour after single-dose i.p. administration. To the best of our knowledge, this is the first time that the spatial distribution of an osmium drug candidate has been investigated using LA-ICP-MS in tissues. Independent measurements of the average ruthenium and osmium concentration via microwave digestion and ICP-MS in organs and tumours were in good agreement with the LA-ICP-MS results. Matrix-matched standards (MMS) ranging from 1 to 30 µg g-1 were prepared to quantify the spatial distributions of the metals and the average metal content of the MMS samples was additionally quantified by ICP-MS after microwave digestion. The recoveries for osmium and ruthenium in the MMS were 105% and 101% on average, respectively, validating the sample preparation procedure of the MMS. Preparation of MMS was carried out under an argon atmosphere to prevent oxidation of osmium-species to the volatile OsO4. The highest metal concentrations were found in the liver, followed by kidney, lung and tumour tissues, while muscles displayed only very low quantities of the respective metal. Both metallodrugs accumulated in the cortex of the kidneys more strongly compared to the medulla. Interestingly, osmium from 2 was largely located at the periphery and tissue edges, whereas ruthenium from 1 was observed to penetrate deeper into the organs and tumours.


Subject(s)
Antineoplastic Agents/administration & dosage , Colonic Neoplasms/drug therapy , Laser Therapy , Mass Spectrometry/methods , Molecular Imaging/methods , Osmium/administration & dosage , Ruthenium/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Mice , Mice, Inbred BALB C , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacokinetics , Osmium/pharmacokinetics , Ruthenium/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
8.
Adv Healthc Mater ; 7(5)2018 03.
Article in English | MEDLINE | ID: mdl-29327505

ABSTRACT

Utilizing the size-dependent adsorption properties of ruthenium carbonyl clusters (Ru-carbon monoxide (CO)) onto graphene oxide (GO), a facile CO-release platform for in situ vasodilation as a treatment for stroke-related vascular diseases is developed. The rate and amount of formation of the CO-release-active RuII (CO)2 species can be modulated by a simple mixing procedure at room temperature. The subsequent thermally induced oxidation of RuII (CO)2 to RuO2 on the GO surface results in the release of CO. Further modulation of thermal and CO-release properties can be achieved via a hybridization of medium- and high-nuclearity of Ru-CO clusters that produces a RuO2 /RuII (CO)2 /6 Ru-CO-GO composite, where 6 Ru-CO-GO provides a photothermally activated reservoir of RuII (CO)2 species and the combined infrared absorption properties of GO and RuO2 provides photothermal response for in situ CO-release. The RuO2 /RuII (CO)2 /6 Ru-CO-GO composite does not produce any cytotoxicity and the efficacy of the composite is further demonstrated in a cortical photothrombotic ischemia rat model.


Subject(s)
Coordination Complexes , Graphite , Hyperthermia, Induced , Phototherapy , Ruthenium , Stroke/therapy , Animals , Cell Line , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/pharmacology , Graphite/chemistry , Graphite/pharmacokinetics , Graphite/pharmacology , Humans , Rats , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology , Stroke/metabolism , Stroke/pathology
9.
Chem Soc Rev ; 46(24): 7706-7756, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29177281

ABSTRACT

Ruthenium(ii) [Ru(ii)] polypyridyl complexes have been the focus of intense investigations since work began exploring their supramolecular interactions with DNA. In recent years, there have been considerable efforts to translate this solution-based research into a biological environment with the intention of developing new classes of probes, luminescent imaging agents, therapeutics and theranostics. In only 10 years the field has expanded with diverse applications for these complexes as imaging agents and promising candidates for therapeutics. In light of these efforts this review exclusively focuses on the developments of these complexes in biological systems, both in cells and in vivo, and hopes to communicate to readers the diversity of applications within which these complexes have found use, as well as new insights gained along the way and challenges that researchers in this field still face.


Subject(s)
Coordination Complexes/pharmacokinetics , Pyridines/pharmacokinetics , Ruthenium/pharmacokinetics , Cell Line , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Humans , Molecular Structure , Pyridines/chemistry , Ruthenium/chemistry
10.
Angew Chem Int Ed Engl ; 56(41): 12628-12633, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28834038

ABSTRACT

Using a new mononuclear "building block," for the first time, a dinuclear RuII (dppn) complex and a heteroleptic system containing both RuII (dppz) and RuII (dppn) moieties are reported. The complexes, including the mixed dppz/dppn system, are 1 O2 sensitizers. However, unlike the homoleptic dppn systems, the mixed dppz/dppn complex also displays a luminescence "switch on" DNA light-switch effect. In both cisplatin sensitive and resistant human ovarian carcinoma lines the dinuclear complexes show enhanced uptake compared to their mononuclear analogue. Thanks to a favorable combination of singlet oxygen generation and cellular uptake properties all three of the new complexes are phototoxic and display potent activity against chemotherapeutically resistant cells.


Subject(s)
Coordination Complexes/pharmacology , Intercalating Agents/pharmacology , Ovarian Neoplasms/drug therapy , Photosensitizing Agents/pharmacology , Ruthenium/pharmacology , Cell Line, Tumor , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , DNA/metabolism , Female , Humans , Intercalating Agents/chemistry , Intercalating Agents/pharmacokinetics , Ovarian Neoplasms/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Singlet Oxygen/metabolism
11.
J Inorg Biochem ; 175: 80-91, 2017 10.
Article in English | MEDLINE | ID: mdl-28738277

ABSTRACT

Two ruthenium(II) complexes Ru1 and Ru2 bearing as a one ligand 2,2'-bipyridine substituted by a semicarbazone 2-formylopyridine moiety (bpySC: 5-(4-{4'-methyl-[2,2'-bipyridine]-4-yl}but-1-yn-1-yl)pyridine-2-carbaldehyde semicarbazone) and as the others 2,2'-bipyridine (bpy) and 4,7-diphenyl-1,10-phenanthroline (dip), respectively, as auxiliary ligands have been prepared. Their biological activity has been studied on murine colon carcinoma (CT26) and human lung adenocarcinoma (A549) cell lines. The anti-proliferative activity was dependent on the presence of bpy or dip in the complex, with one order of magnitude higher cytotoxicity for Ru2 (dip ligands). Ru1 (bpy ligands) exhibited a distinct increase in cytotoxicity going from 24 to 72h of incubation with cells as was not observed for Ru2. Even though both studied compounds were powerful apoptosis inducing agents, the mechanism of their action was entirely different. Ru1-incubated A549 cells showed a notable increase in cells number in the S-phase of the cell cycle, with concomitant decrease in the G2/M phase, while Ru2 promoted a cell accumulation in the G0/G1 phase. In contrast, Ru1 induced marginal oxidative stress in A549 cell lines even upon increasing the incubation time. Even though Ru1 preferably accumulated in lysosomes it triggered the apoptotic cellular death via an intrinsic mitochondrial pathway. Ru1-incubated A549 cells showed swelling and enlarging of the mitochondria. It was not observed in case of Ru2 for which mitochondria and endoplasmic reticulum were found as primarily localization site. Despite this the apoptosis induced by Ru2 was caspase-independent. All these findings point to a pronounced role of auxiliary ligands in tuning the mode of biological activity.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Coordination Complexes , Mitochondria/metabolism , Pyridines , Ruthenium , A549 Cells , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Coordination Complexes/pharmacokinetics , Coordination Complexes/pharmacology , Humans , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium/pharmacology
12.
Sci Rep ; 7(1): 5738, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28720875

ABSTRACT

The constant increase in cardiovascular disease rate coupled with significant drawbacks of existing therapies emphasise the necessity to improve therapeutic strategies. Natural flavonoids exert innumerable pharmacological effects in humans. Here, we demonstrate the effects of chrysin, a natural flavonoid found largely in honey and passionflower on the modulation of platelet function, haemostasis and thrombosis. Chrysin displayed significant inhibitory effects on isolated platelets, however, its activity was substantially reduced under physiological conditions. In order to increase the efficacy of chrysin, a sulfur derivative (thio-chrysin), and ruthenium-complexes (Ru-chrysin and Ru-thio-chrysin) were synthesised and their effects on the modulation of platelet function were evaluated. Indeed, Ru-thio-chrysin displayed a 4-fold greater inhibition of platelet function and thrombus formation in vitro than chrysin under physiologically relevant conditions such as in platelet-rich plasma and whole blood. Notably, Ru-thio-chrysin exhibited similar efficacy to chrysin in the modulation of haemostasis in mice. Increased bioavailability and cell permeability of Ru-thio-chrysin compared to chrysin were found to be the basis for its enhanced activity. Together, these results demonstrate that Ru-thio-coupled natural compounds such as chrysin may serve as promising templates for the development of novel anti-thrombotic agents.


Subject(s)
Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Flavonoids/pharmacology , Hemostasis/drug effects , Platelet Activation/drug effects , Ruthenium/pharmacology , Thrombosis/prevention & control , Animals , Biological Availability , Disease Models, Animal , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/pharmacokinetics , Flavonoids/administration & dosage , Flavonoids/pharmacokinetics , Humans , Mice , Ruthenium/administration & dosage , Ruthenium/pharmacokinetics
13.
Biomaterials ; 129: 111-126, 2017 06.
Article in English | MEDLINE | ID: mdl-28340357

ABSTRACT

Elucidation of the communication between metal complexes and cell membrane may provide useful information for rational design of metal-based anticancer drugs. Herein we synthesized a novel class of ruthenium (Ru) complexes containing phtpy derivatives (phtpy = phenylterpyridine), analyzed their structure-activity relationship and revealed their action mechanisms. The result showed that, the increase in the planarity of hydrophobic Ru complexes significantly enhanced their lipophilicity and cellular uptake. Meanwhile, the introduction of nitro group effectively improved their anticancer efficacy. Further mechanism studies revealed that, complex (2c), firstly accumulated on cell membrane and interacted with death receptors to activate extrinsic apoptosis signaling pathway. The complex was then transported into cell cytoplasm through transferrin receptor-mediated endocytosis. Most of the intracellular 2c accumulated in cell plasma, decreasing the level of cellular ROS, inducing the activation of caspase-9 and thus intensifying the apoptosis. At the same time, the residual 2c can translocate into cell nucleus to interact with DNA, induce DNA damage, activate p53 pathway and enhance apoptosis. Comparing with cisplatin, 2c possesses prolonged circulation time in blood, comparable antitumor ability and importantly, much lower toxicity in vivo. Taken together, this study uncovers the role of membrane receptors in the anticancer actions of Ru complexes, and provides fundamental information for rational design of membrane receptor targeting anticancer drugs.


Subject(s)
Apoptosis/drug effects , Cell Membrane/metabolism , Coordination Complexes/pharmacology , Neoplasms/pathology , Pyridines/pharmacology , Receptors, Death Domain/metabolism , Ruthenium/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Coordination Complexes/pharmacokinetics , DNA Damage , Endocytosis , Humans , Mice, Nude , Models, Biological , Phosphorylation/drug effects , Pyridines/pharmacokinetics , Reactive Oxygen Species/metabolism , Receptors, Transferrin/metabolism , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tissue Distribution/drug effects , Tumor Suppressor Protein p53/metabolism
14.
Chimia (Aarau) ; 71(3): 112-119, 2017 Mar 29.
Article in English | MEDLINE | ID: mdl-28351456

ABSTRACT

Metal-based drugs with novel targets and modes of action are increasingly being developed as alternatives to classical platinum(ii) chemotherapeutics. Imaging methods in tumour cells and tissues offer valuable insights into the behaviour of these novel complexes; however, mapping the distribution of metal ions and complexes within cellular environments remains challenging. The advantages and limitations of three modes of imaging: synchrotron radiation-induced X-ray fluorescence, mass spectrometry, and fluorescence microscopy are discussed in this review, with particular emphasis on their use in imaging ruthenium-based drugs.


Subject(s)
Mass Spectrometry/methods , Microscopy, Fluorescence/methods , Pharmaceutical Preparations/analysis , Ruthenium/pharmacokinetics , Spectrometry, X-Ray Emission/methods , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Humans , Lasers , Pharmaceutical Preparations/chemistry , Ruthenium/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Synchrotrons
15.
Sci Rep ; 7: 41571, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28139763

ABSTRACT

Transition metal sulfide (TMS) holds great potential in cancer photothermal therapy (PTT) because of the high absorbance in the near-infrared (NIR) region. The short blood circulation time and limited tumor accumulation of TMS-based photothermal agents, however, limit their applications. Herein, we design a novel TMS-based PTT agent, ruthenium sulfide-based nanoclusters (NCs), to overcome the current limitations. We firstly develop a simple method to prepare oleic acid coated ruthenium sulfide nanodots (OA-RuS1.7 NDs) and assemble them into water-soluble NCs via sequentially coating with denatured bovine serum albumin (dBSA) and poly(ethylene glycol) (PEG). The obtained PEG-dBSA-RuS1.7 NCs possess excellent photothermal conversion ability. More significantly, they exhibit enhanced blood circulation time and tumor-targeting efficiency in vivo compared with other TMS-based PTT nanoagents, which may be attributed to their appropriate hydrodynamic diameter (~70 nm) and an ideal charge (~0 mV). These characteristics help the PEG-dBSA-RuS1.7 NCs to escape the removal by the reticuloendothelial system (RES) and kidney. All these advantages enable the PEG-dBSA-RuS1.7 NCs to selectively concentrate in tumor sites and effectively ablate the cancer cells upon NIR irradiation.


Subject(s)
Hyperthermia, Induced , Metal Nanoparticles , Neoplasms/metabolism , Neoplasms/therapy , Phototherapy , Ruthenium/metabolism , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Disease Models, Animal , Humans , Hyperthermia, Induced/methods , Metal Nanoparticles/chemistry , Mice , Neoplasms/pathology , Phototherapy/methods , Polyethylene Glycols/chemistry , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
16.
Anticancer Agents Med Chem ; 17(2): 212-220, 2017.
Article in English | MEDLINE | ID: mdl-27039925

ABSTRACT

BACKGROUND: The RAPTA-EA1 complex [ruthenium(II)-arene 1,3,5-triaza-7-phosphaadamantane (pta) complex with an arene-tethered ethacrynic acid ligand] has been reported to overcome drug resistance that developed due to the current use of platinum-based treatments. However, the exact mechanism of action of RAPTA-EA1 remains largely unexplored and unknown. OBJECTIVE: Here we have further studied the effect of RAPTA-EA1 on BRCA1-defective HCC1937 breast cancer cells and compared its effects on BRCA1-competent MCF-7 breast cancer cells. METHOD: HCC1937 and MCF-7 breast cancer cells were treated with the RAPTA-EA1 complex. The cytotoxicity of ruthenium-induced cells was evaluated by a MTT assay. Cellular uptake of ruthenium was determined by ICP-MS. Cell cycle and apoptosis were assessed using a flow cytometer. Expression of BRCA1 mRNA and its encoded protein was quantitated by a real-time RT-PCR and Western blotting. RESULTS: Differences in cytotoxicity were correlated with the differential accumulations of ruthenium and the induction of apoptosis. The ruthenium complex caused dramatically more damage to the BRCA1 gene in the BRCA1-defective HCC1937 cells than to the BRCA1-competent MCF-7 cells. It decreased the expression of BRCA1 mRNA in the BRCA1-competent cells, while in contrast, its expression increased in the BRCA1-defective cells. However, the expression of the BRCA1 protein was significantly reduced in both types of breast cancer cells. CONCLUSION: The results presented here have demonstrated a differential cellular response for the BRCA1-defective and BRCA1-competent breast cancer cells to RAPTA-EA1. These findings have provided more insight into the actions and development of the ruthenium-based compounds for use for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Down-Regulation/drug effects , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, BRCA1/drug effects , Humans , MCF-7 Cells , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , RNA, Messenger/genetics , Ruthenium/chemistry , Ruthenium/pharmacokinetics
17.
Pharmacol Res ; 107: 282-290, 2016 05.
Article in English | MEDLINE | ID: mdl-27038531

ABSTRACT

Ruthenium compounds have become promising alternatives to platinum drugs by displaying specific activities against different cancers and favorable toxicity and clearance properties. Here, we show that the ruthenium(II) complex [Ru(p-cymene)(bis(3,5-dimethylpyrazol-1-yl)methane)Cl]Cl (UNICAM-1) exhibits potent in vivo antitumor effects. When administered as four-dose course, by repeating a single dose (52.4mgkg-1) every three days, UNICAM-1 significantly reduces the growth of A17 triple negative breast cancer cells transplanted into FVB syngeneic mice. Pharmacokinetic studies indicate that UNICAM-1 is rapidly eliminated from kidney, liver and bloodstream thanks to its high hydrosolubility, exerting excellent therapeutic activity with minimal side effects. Immunohistological analysis revealed that the efficacy of UNICAM-1, mainly relies on its capacity to reverse tumor-associated immune suppression by significantly reducing the number of tumor-infiltrating regulatory T cells. Therefore, UNICAM-1 appears very promising for the treatment of TNBC.


Subject(s)
Antineoplastic Agents/therapeutic use , Organometallic Compounds/therapeutic use , Ruthenium/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Organometallic Compounds/blood , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Ruthenium/blood , Ruthenium/pharmacokinetics , Ruthenium/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects
18.
J Antimicrob Chemother ; 71(6): 1547-55, 2016 06.
Article in English | MEDLINE | ID: mdl-26945708

ABSTRACT

OBJECTIVES: The objectives of this study were to: (i) determine the in vitro activities of a series of di-, tri- and tetra-nuclear ruthenium complexes (Rubbn, Rubbn-tri and Rubbn-tetra) against a range of Gram-positive and -negative bacteria and compare the antimicrobial activities with the corresponding toxicities against eukaryotic cells; and (ii) compare MIC values with achievable in vivo serum concentrations for the least toxic ruthenium complex. METHODS: The in vitro activities were determined by MIC assays and time-kill curve experiments, while the toxicities of the ruthenium complexes were determined using the Alamar blue cytotoxicity assay. A preliminary pharmacokinetic study was undertaken to determine the Rubb12 serum concentration in mice as a function of time after administration. RESULTS: Rubb12, Rubb12-tri and Rubb12-tetra are highly active, with MIC values of 1-2 mg/L (0.5-1.5 µM) for a range of Gram-positive strains, but showed variable activities against a panel of Gram-negative bacteria. Time-kill experiments indicated that Rubb12, Rubb12-tri and Rubb12-tetra are bactericidal and kill bacteria within 3-8 h. The di-, tri- and tetra-nuclear complexes were ∼50 times more toxic to Gram-positive bacteria and 25 times more toxic to Gram-negative strains, classified as susceptible, than to liver and kidney cells. Preliminary pharmacokinetic experiments established that serum concentrations higher than MIC values can be obtained for Rubb12 with an administered dose of 32 mg/kg. CONCLUSIONS: The ruthenium complexes, particularly Rubb12, have potential as new antimicrobial agents. The structure of the dinuclear ruthenium complex can be readily further modified in order to increase the selectivity for bacteria over eukaryotic cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Eukaryotic Cells/drug effects , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Colorimetry/methods , Female , Male , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microbial Viability/drug effects , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/toxicity , Oxazines/analysis , Ruthenium/pharmacokinetics , Ruthenium/toxicity , Serum/chemistry , Xanthenes/analysis
19.
Metallomics ; 7(5): 885-95, 2015 May.
Article in English | MEDLINE | ID: mdl-25805368

ABSTRACT

In the present work a novel C,N-cyclometalated benzimidazole Ru(ii) arene complex (GY34) was characterized by applying an alternative, diverse approach considering both chemical and biological aspects. RP-HPLC-ICP-MS and RP-HPLC-ESI-MS analysis proved that GY34 in both RPMI-1640 cell medium and ammonium acetate buffer was transformed into several subspecies and the importance of evaluating and controlling analyte stability throughout experiments was demonstrated. Applying a novel cell fractionation protocol GY34 was found to target cell nuclei and mitochondria in Ehrlich Lettré Ascites (ELA) cells, with the intracellular distribution depending on GY34 concentration in the cell medium during incubation. In ELA cells 96 ± 0.2% of cytosolic GY34 was bound to high-molecular species. Furthermore, using the tracer technique GY34 was found to reduce uptake and increase release of the organic osmolyte taurine in ELA cells, with innate resistance to Cisplatin and in A2780 human ovarian cancer cells, with acquired resistance to Cisplatin. Importantly, FACS analysis revealed that GY34 induced apoptosis in ELA cells. The present data suggest the potential of GY34 in overcoming Cisplatin resistance. The methodology applied can be used as a general protocol and an additional tool in the initial evaluation of novel metal-based drugs.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Female , Homeostasis/drug effects , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ruthenium/pharmacokinetics , Taurine/metabolism
20.
Invest New Drugs ; 33(1): 201-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25344453

ABSTRACT

BACKGROUND: This phase I/II study determined the maximal tolerable dose, dose limiting toxicities, antitumor activity, the pharmacokinetics and pharmacodynamics of ruthenium compound NAMI-A in combination with gemcitabine in Non-Small Cell Lung Cancer patients after first line treatment. METHODS: Initial dose escalation of NAMI-A was performed in a 28 day cycle: NAMI-A as a 3 h infusion through a port-a-cath at a starting dose of 300 mg/m(2) at day 1, 8 and 15, in combination with gemcitabine 1,000 mg/m(2) at days 2, 9 and 16. Subsequently, dose escalation of NAMI-A in a 21 day schedule was explored. At the maximal tolerable dose level of this schedule an expansion group was enrolled of which 15 patients were evaluable for response. RESULTS: Due to frequent neutropenic dose interruptions in the third week, the 28 day schedule was amended into a 21 day schedule. The maximal tolerable dose was 300 and 450 mg/m(2) of NAMI-A (21 day schedule). Main adverse events consisted of neutropenia, anemia, elevated liver enzymes, transient creatinine elevation, nausea, vomiting, constipation, diarrhea, fatigue, and renal toxicity. CONCLUSION: NAMI-A administered in combination with gemcitabine is only moderately tolerated and less active in NSCLC patients after first line treatment than gemcitabine alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Dimethyl Sulfoxide/administration & dosage , Dimethyl Sulfoxide/adverse effects , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacokinetics , Female , Humans , Lung Neoplasms/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Organometallic Compounds/administration & dosage , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacokinetics , Ruthenium/administration & dosage , Ruthenium/adverse effects , Ruthenium/blood , Ruthenium/pharmacokinetics , Ruthenium Compounds , Treatment Outcome , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...