Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
ChemMedChem ; 17(20): e202200444, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36041073

ABSTRACT

Herein we illustrate the formation and characterization of new paramagnetic ruthenium compounds, trans-P-[RuCl(PPh3 )2 (pmt)]Cl (1) (Hpmt=1-((pyridin-2-yl)methylene)thiosemicarbazide), trans-P-[RuCl(PPh3 )2 (tmc)]Cl (2) (Htmc=1-((thiophen-2-yl)methylene)thiosemicarbazide) and a diamagnetic ruthenium complex, cis-Cl, trans-P-[RuCl2 (PPh3 )2 (btm)] (3) (btm=2-((5-hydroxypentylimino)methyl)benzothiazole). Agarose gel electrophoresis experiments of the metal compounds illustrated dose-dependent binding to gDNA by 1-3, while methylene blue competition assays suggested that 1 and 2 are also DNA intercalators. Assessment of the effects of the compounds on topoisomerase function indicated that 1-3 are capable of inhibiting topoisomerase I activity in terms of the ability to nick supercoiled plasmid DNA. The cytotoxic activities of the metal complexes were determined against a range of cancer cell lines versus a non-tumorigenic control cell line, and the complexes were, in general, more cytotoxic towards the cancer cells, displaying IC50 values in the low micromolar range. Time-dependent stability studies showed that in the presence of strong nucleophilic species (such as DMSO), the chloride co-ligands of 1-3 are rapidly substituted by the former as proven by the suppression of the substitution reactions in the presence of an excess amount of chloride ions. The metal complexes are significantly stable in both DCM and an aqueous phosphate buffer containing 2 % DMSO.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Organometallic Compounds , Ruthenium , Thiosemicarbazones , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Ruthenium/pharmacology , Ruthenium/chemistry , Thiosemicarbazones/pharmacology , Coordination Complexes/toxicity , Coordination Complexes/chemistry , Schiff Bases/pharmacology , Dimethyl Sulfoxide , Methylene Blue , Intercalating Agents , Chlorides , DNA Topoisomerases, Type I/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , DNA/chemistry , Benzothiazoles/pharmacology , Phosphates , Organometallic Compounds/chemistry
2.
Inorg Chem ; 60(12): 8826-8837, 2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34060309

ABSTRACT

How to deliver nitric oxide (NO) to a physiological target and control its release quantitatively is a key issue for biomedical applications. Here, a water-soluble nitrosylruthenium complex, [(CH3)4N][RuCl3(5cqn)(NO)] (H5cqn = 5-chloro-8-quinoline), was synthesized, and its structure was confirmed with 1H NMR and X-ray crystal diffraction. Photoinduced NO release was investigated with time-resolved Fourier transform infrared and electron paramagnetic resonance (EPR) spectroscopies. The binding constant of the [RuCl3(5cqn)(NO)]- complex with human serum albumin (HSA) was determined by fluorescence spectroscopy, and the binding mode was identified by X-ray crystallography of the HSA and Ru-NO complex adduct. The crystal structure reveals that two molecules of the Ru-NO complex are located in the subdomain IB, which is one of the major drug binding regions of HSA. The chemical structures of the Ru complexes were [RuCl3(5cqn)(NO)]- and [RuCl3(Glycerin)NO]-, in which the electron densities for all ligands to Ru are unambiguously identified. EPR spin-trapping data showed that photoirradiation triggered NO radical generation from the HSA complex adduct. Moreover, the near-infrared image of exogenous NO from the nitrosylruthenium complex in living cells was observed using a NO-selective fluorescent probe. This study provides a strategy to design an appropriate delivery system to transport NO and metallodrugs in vivo for potential applications.


Subject(s)
Coordination Complexes/metabolism , Nitric Oxide/metabolism , Ruthenium Compounds/metabolism , Serum Albumin, Human/metabolism , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Crystallography, X-Ray , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Nitric Oxide/chemistry , Optical Imaging , Photochemical Processes , Ruthenium Compounds/chemistry , Serum Albumin, Human/chemistry , Tumor Cells, Cultured
3.
ACS Appl Mater Interfaces ; 13(17): 19572-19580, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33900720

ABSTRACT

Photoresponsive ruthenium (Ru) complexes have been extensively studied in the photodynamic therapy (PDT) of cancer. The metal-to-ligand charge transfer (MLCT) absorption maximum of most Ru complexes is located in the short-wavelength visible region, which is well suited for superficial tumors but shows inefficient therapeutic effects for more deep-seated ones. Moreover, Ru complexes are primarily located in the mitochondria or nucleus, always resulting in high levels of dark toxicity and DNA mutation. Herein, we reported a new ruthenium complex (Ru-I) for red-light-triggered PDT. The activation wavelength of Ru-I is successfully extended to 660 nm. Importantly, the complex photosensitizer can be quickly taken up by cancer cells and selectively accumulated in the lysosome, an ideal localization for PDT purposes. Intratumoral injection of Ru-I into tumor-bearing mice achieved excellent therapeutic effects and thus holds great promise for applications in lysosome localization photodynamic therapy.


Subject(s)
Coordination Complexes/pharmacology , Light , Lysosomes/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacology , Ruthenium Compounds/pharmacology , Animals , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Photosensitizing Agents/metabolism , Ruthenium Compounds/metabolism , Subcellular Fractions/metabolism , Xenograft Model Antitumor Assays
4.
Chembiochem ; 22(4): 679-685, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33026156

ABSTRACT

Directed evolution of Cp*RhIII -linked nitrobindin (NB), a biohybrid catalyst, was performed based on an in vitro screening approach. A key aspect of this effort was the establishment of a high-throughput screening (HTS) platform that involves an affinity purification step employing a starch-agarose resin for a maltose binding protein (MBP) tag. The HTS platform enables efficient preparation of the purified MBP-tagged biohybrid catalysts in a 96-well format and eliminates background influence of the host E. coli cells. Three rounds of directed evolution and screening of more than 4000 clones yielded a Cp*RhIII -linked NB(T98H/L100K/K127E) variant with a 4.9-fold enhanced activity for the cycloaddition of acetophenone oximes with alkynes. It is confirmed that this HTS platform for directed evolution provides an efficient strategy for generating highly active biohybrid catalysts incorporating a synthetic metal cofactor.


Subject(s)
Chromatography, Affinity/methods , Chromatography, Agarose/methods , High-Throughput Screening Assays/methods , Maltose-Binding Proteins/metabolism , Organometallic Compounds/metabolism , Ruthenium Compounds/metabolism , Starch/chemistry , Catalysis , Organometallic Compounds/chemistry , Ruthenium Compounds/chemistry
5.
Photochem Photobiol ; 96(2): 327-339, 2020 03.
Article in English | MEDLINE | ID: mdl-31691282

ABSTRACT

A series of strained Ru(II) complexes were studied for potential anticancer activity in hypoxic tissues. The complexes were constructed with methylated ligands that were photolabile and an imidizo[4,5-f][1,10]phenanthroline ligand that contained an appended aromatic group to potentially allow for contributions of ligand-centered excited states. A systematic variation of the size and energy of the aromatic group was performed using systems containing 1-4 fused rings, and the photochemical and photobiological behaviors of all complexes were assessed. The structure and nature of the aromatic group had a subtle impact on photochemistry, altering environmental sensitivity, and had a significant impact on cellular cytotoxicity and photobiology. Up to 5-fold differences in cytotoxicity were observed in the absence of light activation; this rose to 50-fold differences upon exposure to 453 nm light. Most significantly, one complex retained activity under conditions with 1% O2 , which is used to induce hypoxic changes. This system exhibited a photocytotoxicity index (PI) of 15, which is in marked contrast to most other Ru(II) complexes, including those designed for O2 -independent mechanisms of action.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia , Ruthenium Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Coordination Complexes/chemistry , Oxygen/metabolism , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Spectrum Analysis/methods
6.
IEEE Trans Biomed Eng ; 66(11): 3080-3087, 2019 11.
Article in English | MEDLINE | ID: mdl-30794501

ABSTRACT

OBJECTIVE: In this paper, a new method was established to monitor multichannel neural activity with microelectrode arrays (MEAs) under modulation of caged compounds in a rat model of seizures. METHODS: The 16-channel MEAs were fabricated and implanted into the hippocampus of normal rats and epileptic rats for neural spike and local field potential (LFP) recording. Using optical fibers with drug delivery tubing, two different caged compounds [ruthenium-bipyridine-trimethylphosphine glutamate (RuBi-Glu) and ruthenium-bipyridine-trimethylphosphine gamma aminobutyric acid (RuBi-GABA)] were applied, and blue light (465 nm) was used to modulate neural activity. RESULTS: In normal rats, RuBi-Glu excited neural activity, and RuBi-GABA inhibited neural activity. The amplitude of spikes increased 26% from 154 to 194 µV with RuBi-Glu modulation. During RuBi-GABA modulation, spikes recovered to 142 µV. The firing rate increased from 1.4 to 4.5 Hz with RuBi-Glu modulation and decreased to 0.8 Hz after RuBi-GABA modulation. The power of LFPs increased from 566 to 1128 µW with RuBi-Glu modulation and recovered to 710 µW with RuBi-GABA modulation. In epileptic rats, the neural activity during seizures was significantly inhibited by RuBi-GABA modulation. The amplitude of spikes was 242 µV during seizures and decreased to 112 µV with RuBi-GABA modulation. The firing rate decreased from 20.29 to 1.33 Hz with RuBi-GABA modulation. CONCLUSION: Using MEAs, the modulation of neural activity with caged compound photolysis was observed with high temporal-spatial resolution in normal and epileptic rats. SIGNIFICANCE: This new method is important for monitoring neural activity with photo-switchable modulation.


Subject(s)
Action Potentials , Electrophysiology/methods , Hippocampus , Neurophysiology/methods , Seizures/metabolism , Action Potentials/drug effects , Action Potentials/radiation effects , Animals , Electrophysiology/instrumentation , Glutamic Acid/analogs & derivatives , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/radiation effects , Microelectrodes , Neurophysiology/instrumentation , Organometallic Compounds/metabolism , Organometallic Compounds/pharmacology , Photic Stimulation , Photolysis , Rats , Ruthenium Compounds/metabolism , Ruthenium Compounds/pharmacology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
7.
Bioelectrochemistry ; 123: 62-69, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29727765

ABSTRACT

Fungal FAD-dependent glucose dehydrogenases (FADGDHs) are considered to be superior enzymes for glucose sensor strips because of their insensitivity to oxygen and maltose. One highly desirable mediator for enzyme sensor strips is hexaammineruthenium(III) chloride because of its low redox potential and high storage stability. However, in contrast to glucose oxidase (GOx), fungal FADGDH cannot utilize hexaammineruthenium(III) as electron acceptor. Based on strategic structure comparison between FADGDH and GOx, we constructed a mutant of Aspergillus flavus-derived FADGDH, capable of utilizing hexaammineruthenium(III) as electron acceptor: AfGDH-H403D. In AfGDH-H403D, a negative charge introduced at the pathway-entrance leading to the FAD attracts the positively charged hexaammineruthenium(III) and guides it into the pathway. The corresponding amino acid in wild-type GOx is negatively charged, which explains the ability of GOx to utilize hexaammineruthenium(III) as electron acceptor. Electrochemical measurements showed a response current of 46.0 µA for 10 mM glucose with AfGDH-H403D and hexaammineruthenium(III), similar to that with wild-type AfGDH and ferricyanide (47.8 µA). Therefore, AfGDH-H403D is suitable for constructing enzyme electrode strips with hexaammineruthenium(III) chloride as sole mediator. Utilization of this new, improved fungal FADGDH should lead to the development of sensor strips for blood glucose monitoring with increased accuracy and less stringent packing requirements.


Subject(s)
Aspergillus flavus/enzymology , Flavin-Adenine Dinucleotide/metabolism , Glucose 1-Dehydrogenase/metabolism , Ruthenium Compounds/metabolism , Amino Acid Substitution , Aspergillus flavus/genetics , Aspergillus flavus/metabolism , Electrochemical Techniques , Electrons , Glucose 1-Dehydrogenase/genetics , Models, Molecular , Protein Engineering
8.
Met Ions Life Sci ; 182018 02 05.
Article in English | MEDLINE | ID: mdl-29394025

ABSTRACT

There has been much recent interest in the development of therapeutic transition metal-based complexes in part fueled by the clinical success of the platinum(II) anticancer drug, cisplatin. Yet known platinum drugs are limited by their high toxicity, severe side-effects, and incidences of drug resistance. Organometallic ruthenium-arene complexes have risen to prominence as a pharmacophore due to the success of other ruthenium drug candidates in clinical trials. In this chapter, we highlight higher order multinuclear ruthenium-arene complexes and their respective investigations as chemotherapeutic agents. We discuss their unique structural properties and the associated biochemical evaluation in the context of anticancer drug design. We also review the structural considerations for the design of these scaffolds and new therapeutic applications that are uncovered for this class of complexes.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Design , Neoplasms/drug therapy , Organometallic Compounds/therapeutic use , Ruthenium Compounds/therapeutic use , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Coordination Complexes , Humans , Models, Molecular , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology , Organometallic Compounds/adverse effects , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Protein Binding , Ruthenium Compounds/adverse effects , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Structure-Activity Relationship
9.
J Inorg Biochem ; 180: 54-60, 2018 03.
Article in English | MEDLINE | ID: mdl-29245062

ABSTRACT

A pair of ruthenium(II) complex enantiomers, Δ- and Λ-[Ru(bpy)2PBIP]2+ {bpy=2,2'-bipyridine, PBIP=2-(4-bromophenyl)imidazo[4,5-f]1,10-phenanthroline} have been synthesized and characterized. The systematic comparative studies between two enantiomers on their DNA binding-behaviors with calf thymus DNA (CT DNA) were carried out by viscosity measurements, spectrophotometric methods and molecular simulation technology. Additional assays were performed to explore the cytotoxicity of the ruthenium(II) enantiomers against tumor cell lines. DNA-binding studies show that both the enantiomers can bind to CT DNA via intercalative mode, and the Δ form binds to CT DNA more strongly than the Λ form does. Molecular simulation further shows that both the two enantiomers intercalate between base pairs of DNA in minor groove, and that the Δ form intercalates into DNA more deeply than the Λ form does. In addition, the cell proliferation assays show that the Δ form induces a greater cytotoxicity than the Λ form on human cervical cancer HeLa cells, which is positive correlated with the results in DNA binding studies and molecular docking, and implies that the DNA binding affinities of ruthenium(II) polypyridyl complexes might be constitute to the part of their anticancer mechanisms.


Subject(s)
Antineoplastic Agents/metabolism , DNA/metabolism , Ruthenium Compounds/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cattle , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Molecular Docking Simulation , Molecular Probes , Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacology , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Stereoisomerism , Viscosity
10.
Proc Natl Acad Sci U S A ; 114(14): E2846-E2851, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28325874

ABSTRACT

The calcium (Ca2+) uniporter of mitochondria is a holocomplex consisting of the Ca2+-conducting channel, known as mitochondrial calcium uniporter (MCU), and several accessory and regulatory components. A previous electrophysiology study found that the uniporter has high Ca2+ selectivity and conductance and this depends critically on the conserved amino acid sequence motif, DXXE (Asp-X-X-Glu) of MCU. A recent NMR structure of the MCU channel from Caenorhabditis elegans revealed that the DXXE forms two parallel carboxylate rings at the channel entrance that seem to serve as the ion selectivity filter, although direct ion interaction of this structural motif has not been addressed. Here, we use a paramagnetic probe, manganese (Mn2+), to investigate ion and inhibitor binding of this putative selectivity filter. Our paramagnetic NMR data show that mutants with a single carboxylate ring, NXXE (Asn-X-X-Glu) and DXXQ (Asp-X-X-Gln), each can bind Mn2+ specifically, whereas in the WT the two rings bind Mn2+ cooperatively, resulting in ∼1,000-fold higher apparent affinity. Ca2+ can specifically displace the bound Mn2+ at the DXXE site in the channel. Furthermore, titrating the sample with the known channel inhibitor ruthenium 360 (Ru360) can displace Mn2+ binding from the solvent-accessible Asp site but not the inner Glu site. The NMR titration data, together with structural analysis of the DXXE motif and molecular dynamics simulation, indicate that the double carboxylate rings at the apex of the MCU pore constitute the ion selectivity filter and that Ru360 directly blocks ion entry into the filter by binding to the outer carboxylate ring.


Subject(s)
Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Calcium/metabolism , Manganese/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/metabolism , Amino Acid Motifs , Binding Sites , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Magnetic Resonance Spectroscopy , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Molecular Dynamics Simulation , Mutation , Ruthenium Compounds/metabolism , Ruthenium Compounds/pharmacology
11.
Nat Commun ; 7: 12538, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27600651

ABSTRACT

The development of transition metal catalysts capable of promoting non-natural transformations within living cells can open significant new avenues in chemical and cell biology. Unfortunately, the complexity of the cell makes it extremely difficult to translate standard organometallic chemistry to living environments. Therefore, progress in this field has been very slow, and many challenges, including the possibility of localizing active metal catalysts into specific subcellular sites or organelles, remain to be addressed. Herein, we report a designed ruthenium complex that accumulates preferentially inside the mitochondria of mammalian cells, while keeping its ability to react with exogenous substrates in a bioorthogonal way. Importantly, we show that the subcellular catalytic activity can be used for the confined release of fluorophores, and even allows selective functional alterations in the mitochondria by the localized transformation of inert precursors into uncouplers of the membrane potential.


Subject(s)
Metals/metabolism , Ruthenium Compounds/metabolism , Transition Elements/metabolism , Catalysis , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Humans , Metals/chemistry , Molecular Structure , Ruthenium Compounds/chemistry , Transition Elements/chemistry
12.
J Photochem Photobiol B ; 160: 142-53, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27107334

ABSTRACT

Two Ru(II) complexes [Ru(phen)2bppp](ClO4)2 (1) and [Ru(phen)27-Br-dppz](ClO4)2 (2) [phen=1,10 phenanthroline, 7-Br-dppz=7-fluorodipyrido[3,2-a:2',3'-c]phenazine, bppp=11-bromo-pyrido[2',3':5,6]pyrazino[2,3-f] [1,10]phenanthroline] have been synthesized and characterized by elemental analysis, ES-MS, (1)H-NMR, (13)C-NMR and IR. The in vitro cytotoxicity of the complexes examined against a panel of cancer cell lines (HeLa, Du145 and A549) by MTT method, both complexes show prominent anticancer activity against various cancer cells. Live cell imaging study and flow cytometric analysis demonstrate that both the complexes 1 and 2 could cross the cell membrane accumulating in the nucleus. Further, flow cytometry experiments showed that the cytotoxic Ru(II) complexes 1 and 2 induced apoptosis of HeLa tumor cell lines. Photo induced DNA cleavage studies have been performed and results indicate that both the complexes efficiently photo cleave pBR322 DNA. The binding properties of two complexes toward CT-DNA were investigated by various optical methods and viscosity measurements. The experimental results suggested that both Ru(II) complexes can intercalate into DNA base pairs. The complexes were docked into DNA-base pairs using the GOLD docking program.


Subject(s)
Apoptosis/drug effects , DNA/metabolism , Ruthenium Compounds/pharmacology , Anti-Infective Agents/pharmacology , Cell Line, Tumor , Flow Cytometry , Humans , Photochemical Processes , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Spectrophotometry, Ultraviolet , Viscosity
13.
Inorg Chem ; 55(4): 1412-22, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26811966

ABSTRACT

Two new dinuclear Ru(II) polypyridyl complexes containing three and ten methylene chains in their bridging linkers are synthesized and characterized. Their calf thymus DNA-binding and plasmid DNA photocleavage behaviors are comparatively studied with a previously reported, six-methylene-containing analog by absorption and luminescence spectroscopy, steady-state emission quenching by [Fe(CN)6](4-), DNA competitive binding with ethidium bromide, DNA viscosity measurements, DNA thermal denaturation, and agarose gel electrophoresis analyses. Theoretical calculations applying the density functional theory (DFT) method for the three complexes are also performed to understand experimentally observed DNA binding properties. The results show that the two complexes partially intercalate between the base pairs of DNA. Cellular uptake and colocalization studies have demonstrated that the complexes could enter HeLa cells efficiently and localize within lysosomes. The in-vitro antitumor activity against HeLa and MCF-7 tumor cells of the complexes are studied by MTT cytotoxic analysis. A new method, high-content analysis (HCA), is also used to assess cytotoxicity, apoptosis and cell cycle arrest of the three complexes. The results show that the lengths of the alkyl linkers could effectively tune their biological properties and that HCA is suitable for rapidly identifying cytotoxicity and can be substituted for MTT assays to evaluate the cell cytotoxicity of chemotherapeutic agents.


Subject(s)
DNA/metabolism , Ruthenium Compounds/metabolism , Drug Screening Assays, Antitumor , Electrophoresis, Agar Gel , HeLa Cells , Humans , In Vitro Techniques , MCF-7 Cells , Photochemical Processes , Ruthenium Compounds/pharmacology
14.
Article in English | MEDLINE | ID: mdl-26143325

ABSTRACT

A new series of azodye ligands 5-chloro-3-hydroxy-4-(aryldiazenyl)pyridin-2(1H)-one (HLn) were synthesized by coupling of 5-chloro-3-hydroxypyridin-2(1H)-one with aniline and its p-derivatives. These ligands and their Ru(III) complexes of the type trans-[Ru(Ln)2(AsPh3)2]Cl were characterized by elemental analyses, IR, (1)H NMR and UV-Visible spectra as well as magnetic and thermal measurements. The molar conductance measurements proved that all the complexes are electrolytes. IR spectra show that the ligands (HLn) acts as a monobasic bidentate ligand by coordinating via the nitrogen atom of the azo group (NN) and oxygen atom of the deprotonated phenolic OH group, thereby forming a six-membered chelating ring and concomitant formation of an intramolecular hydrogen bond. The molecular and electronic structures of the investigated compounds (HLn) were also studied using quantum chemical calculations. The calf thymus DNA binding activity of the ligands (HLn) and their Ru(III) complexes were studied by absorption spectra and viscosity measurements. The mechanism and the catalytic oxidation of benzyl alcohol by trans-[Ru(Ln)2(AsPh3)2]Cl with hydrogen peroxide as co-oxidant were described.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/metabolism , DNA/chemistry , DNA/metabolism , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Animals , Azo Compounds/chemistry , Azo Compounds/metabolism , Benzyl Alcohol/chemistry , Catalysis , Cattle , Electrochemistry , Electron Spin Resonance Spectroscopy , Hydrogen Bonding , Ligands , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Spectrophotometry, Infrared
15.
Sci Rep ; 5: 9157, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25778692

ABSTRACT

Ruthenium-based complexes have emerged as promising antitumor and antimetastatic agents during the past decades. However, the limited understanding of the antimetastatic mechanisms of these agents is a roadblock to their clinical application. Herein, we reported that, RuPOP, a ruthenium polypyridyl complex with potent antitumor activity, was able to effectively inhibit growth and metastasis of MDA-MB-231 cells and synergistically enhance TRAIL-induced apoptosis. The selective intracellular uptake and cytotoxic effect of RuPOP was found associated with transferring receptor (TfR)-mediated endocytosis. Further investigation on intracellular mechanisms reveled that RuPOP notably suppressed FAK-mediated ERK and Akt activation. Pretreatment of cells with ERK inhibitor (U0126) and PI3K inhibitor (LY294002) significantly potentiated the inhibitory effect of RuPOP on cell growth, migration and invasion. Moreover, the alternation in the expression levels of metastatic regulatory proteins, including uPA, MMP-2/-9, and inhibition of VEGF secretion were also observed after RuPOP treatment. These results demonstrate the inhibitory effect of RuPOP on the growth and metastasis of cancer cells and the enhancement of TRAIL-induced apoptosis though suppression of FAK-mediated signaling. Furthermore, RuPOP exhibits the potential to be developed as a metal-based antimetastatic agent and chemosensitizer of TRAIL for the treatment of human metastatic cancers.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Ruthenium Compounds/pharmacology , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Biological Transport , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Endocytosis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Transferrin/metabolism , Ruthenium Compounds/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
J Am Soc Mass Spectrom ; 25(4): 662-72, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24488754

ABSTRACT

The binding sites of two ruthenium(II) organometallic complexes of the form [(η(6)-arene)Ru(N,N)Cl](+), where arene/N,N = biphenyl (bip)/bipyridine (bipy) for complex AH076, and biphenyl (bip)/o-phenylenediamine (o-pda) for complex AH078, on the peptides angiotensin and bombesin have been investigated using Fourier transform ion cyclotron resonance (FTICR) mass spectrometry. Fragmentation was performed using collisionally activated dissociation (CAD), with, in some cases, additional data being provided by electron capture dissociation (ECD). The primary binding sites were identified as methionine and histidine, with further coordination to phenylalanine, potentially through a π-stacking interaction, which has been observed here for the first time. This initial peptide study was expanded to investigate protein binding through reaction with insulin, on which the binding sites proposed are histidine, glutamic acid, and tyrosine. Further reaction of the ruthenium complexes with the oxidized B chain of insulin, in which two cysteine residues are oxidized to cysteine sulfonic acid (Cys-SO3H), and glutathione, which had been oxidized with hydrogen peroxide to convert the cysteine to cysteine sulfonic acid, provided further support for histidine and glutamic acid binding, respectively.


Subject(s)
Antineoplastic Agents/chemistry , Mass Spectrometry/methods , Organometallic Compounds/chemistry , Peptides/chemistry , Ruthenium Compounds/chemistry , Amino Acid Sequence , Antineoplastic Agents/metabolism , Binding Sites , Molecular Sequence Data , Organometallic Compounds/metabolism , Peptides/metabolism , Protein Binding , Ruthenium Compounds/metabolism
17.
Article in English | MEDLINE | ID: mdl-23994681

ABSTRACT

Three complexes of the general formula trans/cis-[Ru((II))(dppme)(N-N)Cl2] {dppme is H2C=C(CH2PPh2)2 and N-N is 1,2-diaminocyclohexane (trans/cis-(1)) and 1-methyl-1,2-diaminopropane (trans-(2)} were obtained by reacting trans-[RuCl2(dppme)2] with an excess amount of corresponding diamine in CH2Cl2 as a solvent. The complexes were characterized by an elemental analysis, IR, (1)H, (13)C and (31)P{1H} NMR, FAB-MS and UV-visible. The trans-(1) (kinetic product) readily isomerizes to the cis-(1) (thermodynamic product) and this process was followed by using (31)P{(1)H} NMR, cyclic voltammetry and UV-vis spectroscopy. The electrochemical studies on complex (1) reveal that the Ru(III)/Ru(II) couples are sensitive to the isomer (trans/cis) formed. The cis-(1) was confirmed by X-ray structure and (31)P{(1)H} NMR. Transfer-hydrogenation reactions for reduction of trans-4-phenyl-3-butene-2-one were conducted using complexes trans/cis-(1) and trans-(2). The electronic spectra of cis/trans-(1) in dichloromethane were calculated with the use of time-dependent DFT methods.


Subject(s)
Coordination Complexes/chemistry , Coordination Complexes/metabolism , Diamines/chemistry , Ketones/chemistry , Ruthenium Compounds/chemistry , Ruthenium Compounds/metabolism , Catalysis , Crystallography, X-Ray , Electrochemistry , Hydrogenation , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Spectrophotometry, Ultraviolet , Stereoisomerism , Thermodynamics
18.
J Inorg Biochem ; 128: 1-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23911565

ABSTRACT

Prion disease is a neurodegenerative disorder that can occur among humans and other animals. The aberrant isoform of prion protein PrP(Sc) has been identified as the infectious agent. The neuropeptide PrP106-126 has been widely used as a suitable model to study the biological and physiochemical properties of PrP(Sc). PrP106-126 shares several physicochemical and biological properties with PrP(Sc), including cellular toxicity, fibrillogenesis, and membrane-binding affinity. Ruthenium complexes are commonly employed in anti-cancer studies due to their low cellular toxicity. In this study, six hexacoordinated ruthenium complexes with different molecular configurations were used to investigate their effects on PrP106-126 aggregation inhibition. Results revealed that the interaction between the complexes and the peptide included metal coordination and hydrophobic interaction mainly. Those complexes with aromatic structure displayed better inhibitory effects, although they only had a common binding affinity to PrP106-126. This study provided better understanding on the interaction of metal complexes with PrP106-126 and paved the way for potential Ru-based metallodrugs against prion diseases.


Subject(s)
Coordination Complexes/chemistry , Peptide Fragments/chemistry , Prions/chemistry , Ruthenium Compounds/chemistry , Binding, Competitive , Circular Dichroism , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Models, Molecular , Molecular Structure , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Prions/antagonists & inhibitors , Prions/metabolism , Protein Binding , Protein Conformation , Ruthenium Compounds/metabolism , Ruthenium Compounds/pharmacology , Spectrometry, Mass, Electrospray Ionization
19.
J Antimicrob Chemother ; 68(12): 2825-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23873648

ABSTRACT

OBJECTIVES: To determine the energy dependency of and the contribution of the membrane potential to the cellular accumulation of the dinuclear complexes [{Ru(phen)2}2{µ-bbn}](4+) (Rubbn) and the mononuclear complexes [Ru(Me4phen)3](2+) and [Ru(phen)2(bb7)](2+) in Staphylococcus aureus and Escherichia coli, and to examine their effect on the bacterial membrane. METHODS: The accumulation of the ruthenium complexes in bacteria was determined using flow cytometry at a range of temperatures. The cellular accumulation of the ruthenium complexes was also determined in cells that had been incubated with the metal complexes in the presence or absence of metabolic stimulators or inhibitors and/or commercial dyes to determine the membrane potential or membrane permeability. RESULTS: The accumulation of ruthenium complexes in the two bacterial strains was shown to increase with increasing incubation temperature, with the relative increase in accumulation greater with E. coli, particularly for Rubb12 and Rubb16. No decrease in accumulation was observed for Rubb12 in ATP-inhibited cells. While carbonyl cyanide m-chlorophenyl hydrazone (CCCP) did depolarize the cell membrane, no reduction in the accumulation of Rubb12 was observed; however, all ruthenium complexes, when incubated with S. aureus at concentrations twice their MIC, depolarized the membrane to a similar extent to CCCP. Except for the mononuclear complex [Ru(Me4phen)3](2+), incubation of any of the other ruthenium complexes allowed a greater quantity of the membrane-impermeable dye TO-PRO-3 to be taken up by S. aureus. CONCLUSIONS: The results indicate that the potential new antimicrobial Rubbn complexes enter the cell in an energy-independent manner, depolarize the cell membrane and significantly permeabilize the cellular membrane.


Subject(s)
Anti-Infective Agents/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Ruthenium Compounds/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Cell Membrane Permeability/drug effects , Flow Cytometry , Membrane Potentials/drug effects , Temperature
20.
Med Hypotheses ; 80(6): 841-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23587478

ABSTRACT

Lack of specificity and normal tissue toxicity are the two major limitations faced with most of the anticancer agents in current use. Due to effective biodistribution and multimodal cellular actions, during recent past, ruthenium complexes have drawn much attention as next generation anticancer agents. This is because metal center of ruthenium (Ru) effectively binds with the serum transferrin and due to higher concentration of transferrin receptors on the tumor cells, much of the circulating Ru-transferrin complexes are delivered preferentially to the tumor site. This enables Ru-complexes to become tumor cell specific and to execute their anticancer activities in a somewhat targeted manner. Also, there are evidences to suggest that inhibition of phosphodiesterases leads to increased cyclic guanosine monophosphate (cGMP) level, which in turn can evoke cell cycle arrest and can induce apoptosis in the tumor cells. In addition, phosphodiesterase inhibition led increased cGMP level may act as a potent vasodilator and thus, it is likely to enhance blood flow to the growing tumors in vivo, and thereby it can further facilitate delivery of the drugs/compounds to the tumor site. Therefore, it is hypothesized that tagging PDE inhibitors (PDEis) with Ru-complexes could be a relevant strategy to deliver Ru-complexes-PDEi adduct preferentially to the tumor site. The Ru-complex tagged entry of PDEi is speculated to initially enable the tumor cells to become a preferential recipient of such adducts followed by induction of antitumor activities shown by both, the Ru-complex & the PDEi, resulting into enhanced antitumor activities with a possibility of minimum normal tissue toxicity due to administration of such complexes.


Subject(s)
Drug Delivery Systems/methods , Macromolecular Substances/therapeutic use , Models, Biological , Neoplasms/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Ruthenium Compounds/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cyclic GMP/metabolism , Humans , Macromolecular Substances/metabolism , Phosphodiesterase Inhibitors/metabolism , Ruthenium Compounds/metabolism , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...