Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Anal Methods ; 14(11): 1094-1102, 2022 03 17.
Article En | MEDLINE | ID: mdl-34935794

Recent emergence of FTIR spectromicroscopy (micro-FTIR) as a dynamic spectroscopy for imaging to study biological chemistry has opened new possibilities for investigating in situ drug release, redox chemistry effects on biological molecules, DNA and drug interactions, membrane dynamics, and redox reactions with proteins at the single cell level. Micro-FTIR applied to metallodrugs has been playing an important role since the last decade because of its great potential to achieve more robust and controlled pharmacological effects against several diseases, including cancer. An important aspect in the development of these drugs is to understand their cellular properties, such as uptake, accumulation, activity, and toxicity. In this review, we present the potential application of micro-FTIR and its importance for studying metal-based drugs, highlighting the perspectives of chemistry of living cells. We also emphasise bioimaging, which is of high importance to localize the cellular processes, for a proper understanding of the mechanism of action.


DNA , Metals , Oxidation-Reduction , Proteins , Spectroscopy, Fourier Transform Infrared/methods
2.
J Biol Inorg Chem ; 23(6): 903-916, 2018 08.
Article En | MEDLINE | ID: mdl-29971501

The rational design of anti-cancer agents includes a new approach based on ruthenium complexes that can act as nitric oxide (NO) donor agents against specific cellular targets. One of the most studied classes of those compounds is based on bis(bipyridine) ruthenium fragment and its derivative species. In this work, we present the chemical and cytotoxicity properties against the liver hepatocellular carcinoma cell line HepG2 of cis-[RuII(NO+)Cl(dcbpy)2]2- conjugated to a polyclonal antibody IgG (anti-VDAC) recognizing a cell surface marker. UV-visible bands of the ruthenium complex were assigned with the aid of density functional theory, which also allowed estimation of the structures that explain the biological effects of the ruthenium complex-IgG conjugate. The interaction of cis-[RuII(NO+)Cl(dcbpy)2]3- with mitochondria was evaluated due to the potential of these organelles as anti-cancer targets, and considering they interact with the anti-VDAC antibody. The cytotoxicity of cis-[RuII(NO+)Cl(dcbpy)2]3--anti-VDAC antibody was up to 80% greater in comparison to the free cis-[RuII(NO+)Cl(dcbpy)2]3- complex. We suggest that this effect is due to site-specific interaction of the complex followed by NO release.


Antibodies/metabolism , Carcinoma, Hepatocellular/metabolism , Immunoconjugates/metabolism , Liver Neoplasms/metabolism , Mitochondria, Liver/metabolism , Ruthenium/metabolism , Voltage-Dependent Anion Channels/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Mitochondrial Swelling , Nitric Oxide/metabolism , Rats , Rats, Wistar , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channels/immunology
3.
Eur J Pharmacol ; 726: 57-65, 2014 Mar 05.
Article En | MEDLINE | ID: mdl-24462350

The ischemic stroke cascade is composed of several pathophysiological events, providing multiple targets for pharmacological intervention. JM-20 (3-ethoxycarbonyl-2-methyl-4-(2-nitrophenyl)-4,11-dihydro-1H-pyrido[2,3-b][1,5]benzodiazepine) is a novel hybrid molecule, in which a benzodiazepine portion is covalently linked to a dihydropyridine ring, forming a new chemical entity with potential multisite neuroprotective activity. In the present study, JM-20 prevented PC-12 cell death induced either by glutamate, hydrogen peroxide or KCN-mediated chemical hypoxia. This molecule also protected cerebellar granule neurons from glutamate or glutamate plus pentylenetetrazole-induced damage at very low micromolar concentrations. In rat liver mitochondria, JM-20, at low micromolar concentrations, prevented the Ca2+-induced mitochondrial permeability transition, as assessed by mitochondrial swelling, membrane potential dissipation and organelle release of the pro-apoptotic protein cytochrome c. JM-20 also inhibited the mitochondrial hydrolytic activity of F1F0-ATP synthase and Ca2+ influx. Therefore, JM-20 may be a multi-target neuroprotective agent, promoting reductions in neuronal excitotoxic injury and the protection of the mitochondria from Ca2+-induced impairment as well as the preservation of cellular energy balance.


Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Brain Ischemia/pathology , Dihydropyridines/chemistry , Mitochondria/drug effects , Neurons/drug effects , Neurons/pathology , Niacin/analogs & derivatives , Animals , Brain Ischemia/complications , Calcium/metabolism , Cell Death/drug effects , Cerebellum/cytology , Cytochromes c/metabolism , Glutamic Acid/pharmacology , Hydrogen Peroxide/pharmacology , Hydrolysis/drug effects , Liver/drug effects , Liver/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Swelling/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Niacin/chemistry , Niacin/pharmacology , Oxidative Stress/drug effects , PC12 Cells , Pentylenetetrazole/pharmacology , Phosphates/metabolism , Potassium Cyanide/pharmacology , Rats , Stroke/complications
4.
Nitric Oxide ; 26(3): 174-81, 2012 Mar 31.
Article En | MEDLINE | ID: mdl-22349020

Nitrosyl ruthenium complexes are promising NO donor agents with numerous advantages for the biologic applications of NO. We have characterized the NO release from the nitrosyl ruthenium complex [Ru(NO(2))(bpy)(2)(4-pic)](+) (I) and the reactive oxygen/nitrogen species (ROS/RNS)-mediated NO actions on isolated rat liver mitochondria. The results indicated that oxidation of mitochondrial NADH promotes NO release from (I) in a manner mediated by NO(2) formation (at neutral pH) as in mammalian cells, followed by an oxygen atom transfer mechanism (OAT). The NO released from (I) uncoupled mitochondria at low concentrations/incubation times and inhibited the respiratory chain at high concentrations/incubation times. In the presence of ROS generated by mitochondria NO gave rise to peroxynitrite, which, in turn, inhibited the respiratory chain and oxidized membrane protein-thiols to elicit a Ca(2+)-independent mitochondrial permeability transition; this process was only partially inhibited by cyclosporine-A, almost fully inhibited by the thiol reagent N-ethylmaleimide (NEM) and fully inhibited by the NO scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO). These actions correlated with the release of cytochrome c from isolated mitochondria as detected by Western blotting analysis. These events, typically involved in cell necrosis and/or apoptosis denote a potential specific action of (I) and analogs against tumor cells via mitochondria-mediated processes.


Coordination Complexes/pharmacokinetics , Mitochondria, Liver/metabolism , NADP/metabolism , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide/pharmacokinetics , Ruthenium/pharmacokinetics , Analysis of Variance , Animals , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Cytochromes c/metabolism , Hydrogen-Ion Concentration , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Ruthenium/chemistry , Ruthenium/metabolism , Sulfhydryl Compounds
5.
Redox Rep ; 16(3): 108-13, 2011.
Article En | MEDLINE | ID: mdl-21801492

We have used two different probes with distinct detection properties, dichlorodihydrofluorescein diacetate and Amplex Red/horseradish peroxidase, as well as different respiratory substrates and electron transport chain inhibitors, to characterize the reactive oxygen species (ROS) generation by the respiratory chain in calcium-overloaded mitochondria. Regardless of the respiratory substrate, calcium stimulated the mitochondrial generation of ROS, which were released at both the mitochondrial-matrix side and the extra-mitochondrial space, in a way insensitive to the mitochondrial permeability transition pores inhibitor cyclosporine A. In glutamate/malate-energized mitochondria, inhibition at complex I or complex III (ubiquinone cycle) similarly modulated ROS generation at either mitochondrial-matrix side or extra-mitochondrial space; this also occurred when the backflow of electrons to complex I in succinate-energized mitochondria was inhibited. On the other hand, in succinate-energized mitochondria the modulation of ROS generation at mitochondrial-matrix side or extra-mitochondrial space depends on the site of complex III which was inhibited. These results allow a straight comparison between the effects of different respiratory substrates and electron transport chain inhibitors on ROS generation at either mitochondrial-matrix side or extra-mitochondrial space in calcium-overloaded mitochondria.


Calcium/pharmacology , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Analysis of Variance , Animals , Cyclosporine/pharmacology , Electron Transport , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Glutamic Acid/pharmacology , Hydrogen Peroxide/metabolism , Male , Membrane Potential, Mitochondrial , Mitochondria, Liver/drug effects , Mitochondrial Membranes/metabolism , Oxazines/metabolism , Rats , Rats, Wistar , Rotenone/pharmacology , Succinic Acid/pharmacology
6.
Toxicol Appl Pharmacol ; 253(3): 282-9, 2011 Jun 15.
Article En | MEDLINE | ID: mdl-21549140

Guttiferone-A (GA) is a natural occurring polyisoprenylated benzophenone with cytotoxic action in vitro and anti-tumor action in rodent models. We addressed a potential involvement of mitochondria in GA toxicity (1-25 µM) toward cancer cells by employing both hepatic carcinoma (HepG2) cells and succinate-energized mitochondria, isolated from rat liver. In HepG2 cells GA decreased viability, dissipated mitochondrial membrane potential, depleted ATP and increased reactive oxygen species (ROS) levels. In isolated rat-liver mitochondria GA promoted membrane fluidity increase, cyclosporine A/EGTA-insensitive membrane permeabilization, uncoupling (membrane potential dissipation/state 4 respiration rate increase), Ca²âº efflux, ATP depletion, NAD(P)H depletion/oxidation and ROS levels increase. All effects in cells, except mitochondrial membrane potential dissipation, as well as NADPH depletion/oxidation and permeabilization in isolated mitochondria, were partly prevented by the a NAD(P)H regenerating substrate isocitrate. The results suggest the following sequence of events: 1) GA interaction with mitochondrial membrane promoting its permeabilization; 2) mitochondrial membrane potential dissipation; 3) NAD(P)H oxidation/depletion due to inability of membrane potential-sensitive NADP+ transhydrogenase of sustaining its reduced state; 4) ROS accumulation inside mitochondria and cells; 5) additional mitochondrial membrane permeabilization due to ROS; and 6) ATP depletion. These GA actions are potentially implicated in the well-documented anti-cancer property of GA/structure related compounds.


Antineoplastic Agents, Phytogenic/pharmacology , Benzophenones/pharmacology , Mitochondrial Membranes/drug effects , Oxidative Stress/drug effects , Adenosine Triphosphate/analysis , Animals , Benzophenones/pharmacokinetics , Calcium/metabolism , Cell Survival/drug effects , Energy Metabolism/drug effects , Hep G2 Cells , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Swelling/drug effects , NAD/analysis , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
7.
Mitochondrion ; 11(2): 255-63, 2011 Mar.
Article En | MEDLINE | ID: mdl-21044702

Nemorosone, a natural-occurring polycyclic polyprenylated acylphloroglucinol, has received increasing attention due to its strong in vitro anti-cancer action. Here, we have demonstrated the toxic effect of nemorosone (1-25 µM) on HepG2 cells by means of the MTT assay, as well as early mitochondrial membrane potential dissipation and ATP depletion in this cancer cell line. In mitochondria isolated from rat liver, nemorosone (50-500 nM) displayed a protonophoric uncoupling activity, showing potency comparable to the classic protonophore, carbonyl cyanide m-chlorophenyl hydrazone (CCCP). Nemorosone enhanced the succinate-supported state 4 respiration rate, dissipated mitochondrial membrane potential, released Ca(2+) from Ca(2+)-loaded mitochondria, decreased Ca(2+) uptake and depleted ATP. The protonophoric property of nemorosone was attested by the induction of mitochondrial swelling in hyposmotic K(+)-acetate medium in the presence of valinomycin. In addition, uncoupling concentrations of nemorosone in the presence of Ca(2+) plus ruthenium red induced the mitochondrial permeability transition process. Therefore, nemorosone is a new potent protonophoric mitochondrial uncoupler and this property is potentially involved in its toxicity on cancer cells.


Antineoplastic Agents/pharmacology , Benzophenones/pharmacology , Mitochondria, Liver/drug effects , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Humans , Male , Membrane Potentials , Mitochondria, Liver/metabolism , Mitochondrial Swelling/drug effects , Rats , Rats, Wistar
8.
Biochim Biophys Acta ; 1787(3): 176-82, 2009 Mar.
Article En | MEDLINE | ID: mdl-19161974

The oxidation of critical cysteines/related thiols of adenine nucleotide translocase (ANT) is believed to be an important event of the Ca(2+)-induced mitochondrial permeability transition (MPT), a process mediated by a cyclosporine A/ADP-sensitive permeability transition pores (PTP) opening. We addressed the ANT-Cys(56) relative mobility status resulting from the interaction of ANT/surrounding cardiolipins with Ca(2+) and/or ADP by means of computational chemistry analysis (Molecular Interaction Fields and Molecular Dynamics studies), supported by classic mitochondrial swelling assays. The following events were predicted: (i) Ca(2+) interacts preferentially with the ANT surrounding cardiolipins bound to the H4 helix of translocase, (ii) weakens the cardiolipins/ANT interactions and (iii) destabilizes the initial ANT-Cys(56) residue increasing its relative mobility. The binding of ADP that stabilizes the conformation "m" of ANT and/or cardiolipin, respectively to H5 and H4 helices, could stabilize their contacts with the short helix h56 that includes Cys(56), accounting for reducing its relative mobility. The results suggest that Ca(2+) binding to adenine nucleotide translocase (ANT)-surrounding cardiolipins in c-state of the translocase enhances (ANT)-Cys(56) relative mobility and that this may constitute a potential critical step of Ca(2+)-induced PTP opening.


Calcium/metabolism , Cardiolipins/metabolism , Cysteine/metabolism , Mitochondria, Liver/drug effects , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Adenosine Diphosphate/metabolism , Animals , Cell Membrane Permeability/drug effects , Computer Simulation , Cyclosporine/pharmacology , Immunosuppressive Agents/pharmacology , Male , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Models, Chemical , Models, Molecular , Oxidative Stress , Protein Conformation , Rats , Rats, Wistar , Succinic Acid/pharmacology
9.
Nitric Oxide ; 20(1): 24-30, 2009 Feb.
Article En | MEDLINE | ID: mdl-18950724

The ruthenium nitrosyl complex trans-[Ru(NO)(NH(3))(4)(py)](PF(6))(3) (pyNO), a nitric oxide (NO) donor, was studied in regard to the release of NO and its impact both on isolated mitochondria and HepG2 cells. In isolated mitochondria, NO release from pyNO was concomitant with NAD(P)H oxidation and, in the 25-100 microM range, it resulted in dissipation of mitochondrial membrane potential, inhibition of state 3 respiration, ATP depletion and reactive oxygen species (ROS) generation. In the presence of Ca(2+), mitochondrial permeability transition (MPT), an unspecific membrane permeabilization involved in cell necrosis and some types of apoptosis, was elicited. As demonstrated by externalization of phosphatidylserine and activation of caspase-9 and caspase-3, pyNO (50-100 microM) induced HepG2 cell death, mainly by apoptosis. The combined action of the NO itself, the peroxynitrite yielded by NO in the presence of reactive oxygen species (ROS) and the oxidative stress generated by the NAD(P)H oxidation is proposed to be involved in cell death by pyNO, both via respiratory chain inhibition and ROS levels increase, or even via MPT, if Ca(2+) is present.


Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , NADPH Oxidases/metabolism , Oxidation-Reduction , Rats , Reactive Oxygen Species/metabolism
...