Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 13(37): 43892-43903, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34516076

ABSTRACT

Despite technological advancement, nosocomial infections are prevalent due to the rise of antibiotic resistance. A combinatorial approach with multimechanistic antibacterial activity is desired for an effective antibacterial medical device surface strategy. In this study, an antimicrobial peptide, nisin, is immobilized onto biomimetic nitric oxide (NO)-releasing medical-grade silicone rubber (SR) via mussel-inspired polydopamine (PDA) as a bonding agent to reduce the risk of infection. Immobilization of nisin on NO-releasing SR (SR-SNAP-Nisin) and the surface characteristics were characterized by Fourier transform infrared spectroscopy and scanning electron microscopy with energy-dispersive X-ray spectroscopy and contact angle measurements. The NO release profile (7 days) and diffusion of SNAP from SR-SNAP-Nisin were quantified using chemiluminescence-based nitric oxide analyzers and UV-vis spectroscopy, respectively. Nisin quantification showed a greater affinity of nisin immobilization toward SNAP-doped SR. Matrix-assisted laser desorption/ionization mass spectrometry analysis on surface nisin leaching for 120 h under physiological conditions demonstrated the stability of nisin immobilization on PDA coatings. SR-SNAP-Nisin shows versatile in vitro anti-infection efficacy against Gram-negative Escherichia coli and Gram-positive Staphylococcus aureus in the planktonic and adhered states. Furthermore, the combination of NO and nisin has a superior ability to impair biofilm formation on polymer surfaces. SR-SNAP-Nisin leachates did not elicit cytotoxicity toward mouse fibroblast cells and human umbilical vein endothelial cells, indicating the biocompatibility of the material in vitro. The preventative and therapeutic potential of SR-SNAP-Nisin dictated by two bioactive agents may offer a promising antibacterial surface strategy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Immobilized Proteins/pharmacology , Nisin/pharmacology , Nitric Oxide Donors/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Biofilms/drug effects , Cell Survival/drug effects , Escherichia coli/drug effects , Escherichia coli/physiology , Immobilized Proteins/chemistry , Immobilized Proteins/toxicity , Indoles/chemistry , Indoles/toxicity , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Nisin/chemistry , Nisin/toxicity , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/toxicity , Polymers/chemistry , Polymers/toxicity , S-Nitroso-N-Acetylpenicillamine/chemistry , S-Nitroso-N-Acetylpenicillamine/toxicity , Silicone Elastomers/chemistry , Silicone Elastomers/toxicity , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology
2.
ACS Appl Mater Interfaces ; 12(18): 20158-20171, 2020 May 06.
Article in English | MEDLINE | ID: mdl-32271542

ABSTRACT

The management of thrombosis and bacterial infection is critical to ensure the functionality of medical devices. While administration of anticoagulants is the current antithrombotic clinical practice, a variety of complications, such as uncontrolled hemorrhages or heparin-induced thrombocytopenia, can occur. Additionally, infection rates remain a costly and deadly complication associated with use of these medical devices. It has been hypothesized that if a synthetic surface could mimic the biochemical mechanisms of the endothelium of blood vessels, thrombosis could be reduced, anticoagulant use could be avoided, and infection could be prevented. Herein, the interfacial biochemical effects of the endothelium were mimicked by altering the surface of medical grade silicone rubber (SR). Surface modification was accomplished via heparin surface immobilization (Hep) and the inclusion of a nitric oxide (NO) donor into the SR polymeric matrix to achieve synergistic effects (Hep-NO-SR). An in vitro bacteria adhesion study revealed that Hep-NO-SR exhibited a 99.46 ± 0.17% reduction in viable bacteria adhesion compared to SR. An in vitro platelet study revealed Hep-NO-SR reduced platelet adhesion by 84.12 ± 6.19% compared to SR, while not generating a cytotoxic response against fibroblast cells. In a 4 h extracorporeal circuit model without systemic anticoagulation, all Hep-NO-SR samples were able to maintain baseline platelet count and device patency; whereas 66% of SR samples clotted within the first 2 h of study. Results indicate that Hep-NO-SR creates a more hemocompatible and antibacterial surface by mimicking two key biochemical functions of the native endothelium.


Subject(s)
Biomimetic Materials/chemistry , Hematologic Agents/therapeutic use , Heparin/therapeutic use , Nitric Oxide Donors/therapeutic use , S-Nitroso-N-Acetylpenicillamine/therapeutic use , Animals , Bacterial Adhesion/drug effects , Biomimetic Materials/toxicity , Blood Coagulation/drug effects , Blood Platelets/metabolism , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/toxicity , Endothelium/chemistry , Hematologic Agents/pharmacology , Hematologic Agents/toxicity , Heparin/pharmacology , Heparin/toxicity , Immobilized Proteins/pharmacology , Immobilized Proteins/therapeutic use , Immobilized Proteins/toxicity , Mice , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/toxicity , Platelet Adhesiveness/drug effects , Rabbits , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/toxicity , Silicone Elastomers/chemistry , Silicone Elastomers/toxicity , Staphylococcus aureus/drug effects , Surface Properties
3.
Clin Exp Ophthalmol ; 43(6): 568-77, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25801048

ABSTRACT

BACKGROUND: Oxidative and nitrative processes have an important role in the pathogenesis of glaucomatous neurodegeneration. Oxidative stress occurs when cellular production of reactive oxygen species outweighs the protective capacity of antioxidant defences. Reactive oxygen species are generated as by-products of cellular metabolism, primarily in the mitochondria. Herein, we present a novel investigation of the effects of molecular hydrogen (H2 ) on retinal cells exposed to oxidative stress. METHODS: We cultured adult rat retinal tissues in an organotypic culture system with a nitric oxide donor, S-nitroso-N-acetylpenicillamine, in the presence or absence of H2 . Loss of mitochondrial membrane potential and apoptosis of retinal cells were analysed using a MitoTMRE detection kit and TdT-mediated dUTP nick end labeling (TUNEL) assay, respectively. Tyrosine nitration levels and oxidative stress damage in the retina were evaluated using immunohistochemical staining. Retinal damage was quantified by measuring the numbers of cells in the ganglion cell and inner nuclear layers and the thickness of the retina. RESULTS: H2 suppressed loss of mitochondrial membrane potential and apoptosis in retinal cells. Moreover, H2 decreased the tyrosine nitration level and suppressed oxidative stress damage in retinal cells. S-nitroso-N-acetylpenicillamine treatment decreased the cell numbers in the ganglion cell layer and inner nuclear layer, but the presence of H2 inhibited this reduction. These findings suggest that H2 has a neuroprotective effect against retinal cell oxidative damage, presumably by scavenging peroxynitrite. CONCLUSIONS: H2 reduces cellular peroxynitrite, a highly toxic reactive nitrogen species. Thus, H2 may be an effective and novel clinical tool for treating glaucoma and other oxidative stress-related diseases.


Subject(s)
Hydrogen/pharmacology , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Peroxynitrous Acid/toxicity , Retina/drug effects , Animals , Apoptosis/drug effects , Cell Survival , In Situ Nick-End Labeling , Male , Membrane Potential, Mitochondrial/drug effects , Nitrosation , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Retina/metabolism , Retina/pathology , S-Nitroso-N-Acetylpenicillamine/toxicity , Tyrosine/metabolism
4.
Brain Res ; 1543: 308-14, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24275198

ABSTRACT

Genipin, the multipotent ingredient in Gardenia jasmenoides fruit extract (GFE), may be an effective candidate for treatment following stroke or traumatic brain injury (TBI). Secondary injury includes damage mediated by reactive oxygen species (ROS) and reactive nitrogen species (RNS), which can alter the biological function of key cellular structures and eventually lead to cell death. In this work, we studied the neuroprotective potential of genipin against damage stemming from ROS and RNS production in organotypic hippocampal slice cultures (OHSC), as well as its potential as a direct free radical scavenger. A 50 µM dose of genipin provided significant protection against tert-butyl hydroperoxide (tBHP), a damaging organic peroxide. This dosage of genipin significantly reduced cell death at 48 h compared to vehicle control (0.1% DMSO) when administered 0, 1, 6, and 24 h after addition of tBHP. Similarly, genipin significantly reduced cell death at 48 h when administered 0, 1, 2, and 6h after addition of rotenone, which generates reactive oxygen species via a more physiologically relevant mechanism. Furthermore, genipin significantly reduced both cell death and nitrite levels at 24 h caused by S-nitroso-N-acetylpenicillamine (SNAP), a direct nitric oxide (NO) donor, and successfully quenched 1,1-Diphenyl-2-picryl-hydrazyl (DPPH), a stable free radical, suggesting that genipin may act as a direct free radical scavenger. Our encouraging findings suggest that genipin should be tested in animal models of CNS injury with a significant component of ROS- and RNS-mediated damage, such as TBI and stroke, to assess its in vivo efficacy.


Subject(s)
Hippocampus/drug effects , Hippocampus/injuries , Iridoids/pharmacology , Neuroprotective Agents/pharmacology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Analysis of Variance , Animals , Animals, Newborn , Ascorbic Acid/pharmacology , Biphenyl Compounds/pharmacology , Cell Death/drug effects , Dose-Response Relationship, Drug , Insecticides/toxicity , Nitric Oxide Donors/toxicity , Organ Culture Techniques , Picrates/pharmacology , Rotenone/toxicity , S-Nitroso-N-Acetylpenicillamine/toxicity , Time Factors
5.
Cell Biol Toxicol ; 28(3): 161-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22453795

ABSTRACT

The perception of toxicity to nitric oxide (NO) and irradiation (IR) by three different cell types has been studied. The three cell types are the macrophage like RAW264.7 cells, EL4 lymphoma cells, and splenocytes, which represent the different components of a tumor. These three cell types respond differently to NO donors (SNP and SNAP) and radiation treatment. The macrophages were found to be most radio-resistant and insensitive to NO donors. The innate resistance of the macrophages was not due to its antioxidant defense system since there was no significant activation of the enzymes (superoxide dismutases, catalase, and glutathione peroxidase) in RAW264.7 cells after NO donor and irradiation. But the cell cycle arrest of the three cell types was different from each other. The EL4 cells were found to arrest in the G2/M phase while the macrophages were found arrested in the G1 phase of the cell cycle. Such specific killing of the tumor cell in response to NO donor while sparing the macrophages can be of immense importance to radiotherapy.


Subject(s)
Gamma Rays/adverse effects , Macrophages/drug effects , Macrophages/radiation effects , Nitric Oxide/toxicity , Animals , Antioxidants/metabolism , Catalase/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Survival , DNA Fragmentation , Enzyme Activation , Glutathione Peroxidase/metabolism , Macrophages/metabolism , Male , Mice , Nitric Oxide/metabolism , Nitric Oxide Donors/toxicity , Nitroprusside/toxicity , S-Nitroso-N-Acetylpenicillamine/toxicity , Superoxide Dismutase/metabolism , Toxicity Tests/methods
7.
Invest Ophthalmol Vis Sci ; 51(5): 2575-86, 2010 May.
Article in English | MEDLINE | ID: mdl-19959646

ABSTRACT

PURPOSE: To determine whether retinal neurons become more susceptible to injury by nitric oxide (NO) under hypoxic conditions. METHODS: Cells from the RGC-5 line were exposed to different concentrations (0.1-100 microM) of S-nitroso-N-acetyl-penicillamine (SNAP), an NO donor, under normoxic and hypoxic (1.0% O(2)) conditions with 5.5 mM glucose or with no glucose. In some experiments, carboxy-PTIO, a scavenger of NO, was added with SNAP. The SNAP-induced cell injury was determined by the WST-8 assay and by the assessment of phosphatidylserine externalization and changes in hypodiploid DNAs. Alterations of mitochondrial membrane potential, superoxide anion formation, cellular adenosine triphosphate (ATP) contents, and caspase activity were also determined after exposure to SNAP. RESULTS: Exposure of RGC-5 cells to SNAP (100 microM) significantly decreased the number of living cells cultured under hypoxic conditions with or without glucose. Coadministration of carboxy-PTIO (1.0 microM) suppressed SNAP-induced cell death. SNAP-induced cell death of cells cultured under hypoxia with glucose was accompanied by increased expression of phosphatidylserine and hypodiploid DNAs. These findings indicated that death was mediated in part by apoptosis. In addition, loss of mitochondrial membrane potential, increase of superoxide formation, and activation of caspase was observed. Cyclosporine A, TEMPOL, and Z-VAD-FMK suppressed cell death. On the other hand, SNAP depleted the ATP contents of cells cultured under hypoxia without glucose, causing mainly necrotic cell death. CONCLUSIONS: These results indicate that RGC-5 cells become susceptible to SNAP under hypoxic conditions in which NO may have greater impact on mitochondrial function.


Subject(s)
Hypoxia/metabolism , Nitric Oxide Donors/toxicity , Retinal Ganglion Cells/drug effects , S-Nitroso-N-Acetylpenicillamine/toxicity , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Benzoates/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cell Line , DNA/analysis , Diploidy , Enzyme Inhibitors/pharmacology , Flow Cytometry , Glucose/pharmacology , Humans , Imidazoles/pharmacology , Membrane Potential, Mitochondrial/drug effects , Peroxynitrous Acid/metabolism , Phosphatidylserines/metabolism , Retinal Ganglion Cells/metabolism , Superoxides/metabolism
8.
Brain Res ; 1230: 265-72, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18644353

ABSTRACT

Nitric oxide (NO) induces cell proliferation or cell death, depending on the cell type involved, the isoform of nitric oxide synthase activated, and its cellular localisation. In neurons, the damaging effect of NO is usually attributed to the highly toxic peroxynitrite, formed by its reaction with superoxide. Peroxynitrite induces DNA damage and consequently the activation of poly (ADP-ribose) polymerase (PARP). This study set out to examine the contribution of peroxynitrite to the damage induced in cerebellar granule neurons (CGNs) by treatment with the NO donor S-nitroso-N-acetylpenicillamine (SNAP), for short (6 h) or prolonged (24 h) exposures. The Alamar blue assay was used to quantify CGN viability, which was also assessed by morphological examination. SNAP (10 microM-1 mM) induced a concentration- and time-dependent reduction of CGN viability, with associated damage to cell bodies and neurite processes evident following 100 microM SNAP treatments. Damage from 6 h exposures was prevented by the presence of haemoglobin (a NO scavenger), uric acid (a peroxynitrite scavenger), melatonin (a non-specific antioxidant), and by cyclosporin A (a permeability transition pore blocker). It was reduced by the PARP-1 inhibitor 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1(2H)-isoquinolinone (DPQ), whilst superoxide dismutase (SOD) potentiated the effects. Following 24 h exposure to SNAP, damage was only partially blocked by haemoglobin, melatonin, cyclosporin A and DPQ, but was not affected by uric acid or SOD. The data suggest that short exposure to NO induces neuronal damage through peroxynitrite produced by its interaction with superoxide, whereas a longer exposure to NO can induce damage partly by a mechanism which is independent of peroxynitrite formation.


Subject(s)
Cerebellum/pathology , Neurons/pathology , Nitric Oxide Donors/toxicity , Peroxynitrous Acid/metabolism , S-Nitroso-N-Acetylpenicillamine/toxicity , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cyclosporine/pharmacology , Cytoplasmic Granules/pathology , Data Interpretation, Statistical , Free Radical Scavengers/pharmacology , Hemoglobins/metabolism , Hydroquinones/toxicity , Melatonin/pharmacology , Mitochondrial Membranes/drug effects , Neurons/drug effects , Poly Adenosine Diphosphate Ribose/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Uric Acid/pharmacology
9.
Hepatology ; 42(3): 598-607, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116630

ABSTRACT

Nitric oxide (NO) can be either cytoprotective or cytotoxic in hepatocytes, depending on conditions within the cell. We hypothesized that redox status is a determinant of NO effects on cell viability. To cause the disturbance of redox homeostasis in the hepatocytes, cells were treated with the following glutathione (GSH) depleting agents: (1) chronic depletion by 18 hours pretreatment with buthionine sulfoximine (BSO), which depletes GSH by blocking its biosynthesis; and (2) acute depletion by 1 hour pretreatment with diethyl maleate (DEM), which conjugates GSH by the GSH-S-transferase catalyzed reaction. S-nitroso-N-acetyl-D,L-penicillamine (SNAP), a NO donor, was added after removal of GSH-depleting agents. Individual treatment with either SNAP or GSH depletion did not appreciably affect viability. A significant increase of cytotoxicity in hepatocytes was observed with the combination of a concentration and time course regimen of SNAP and GSH depletion. SNAP treatment of GSH-depleted hepatocytes led to an increase in LDH release and oxidative stress, disruption of mitochondrial membrane potential, the presence of nitrotyrosine (an indicator of peroxynitrite (ONOO-) generation), and a decrease in adenosine triphosphate (ATP) content. The interference of mitochondrial respiratory enzymes, especially with the combination treatments, indicated different levels of disturbance of electron transfer, superoxide generation, and ATP production. Other commonly used NO donors were found to exhibit lower and slower toxicity in the setting of GSH depletion than that evident with SNAP. In conclusion, the disruption of cellular redox homeostasis by GSH depletion leads hepatocytes to be more susceptible to NO (especially S-nitrosothiols) and subsequent necrotic cell death.


Subject(s)
Cell Survival/drug effects , Glutathione/deficiency , Hepatocytes/physiology , Nitric Oxide Donors/toxicity , Animals , Cell Membrane Permeability/drug effects , Cells, Cultured , Glutathione/pharmacology , Hepatocytes/drug effects , Hepatocytes/pathology , Kinetics , Rats , Rhodamine 123 , S-Nitroso-N-Acetylpenicillamine/toxicity
10.
Life Sci ; 73(12): 1517-26, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12865091

ABSTRACT

There is growing evidence that high concentrations of nitric oxide (NO), generated by activated astrocytes, might be involved in a variety of neurodegenerative diseases, such as Alzheimer's disease, ischemia and epilepsy. It has recently been suggested that glial cells may produce NO under superoxide radical stimulation by enzyme-independent mechanism. This suggests that also natural antioxidants may have therapeutical relevance in neurodegenerative diseases. Studies of Bhattacharya et al. have evidenced that Bacopa monniera (BM) (family Scrophulariaceae), an Ayurvedic medicinal plant clinically used for memory enhancing, epilepsy, insomnia and as a mild sedative, is able to reduce the memory-dysfunction in rat models of Alzheimer's disease, but the molecular mechanisms of this action are yet to be determined. In the present study, we examined the effect of a methanolic extract of BM on toxicity induced by the nitric oxide donor, S-nitroso-N-acetyl-penicillamine (SNAP), in culture of purified rat astrocytes. Our results indicate that, after 18 h of treatment, SNAP induced an increase in the production of reactive species, but did not induce the rupture of cellular membrane. Conversely, this NO donor induced a fragmentation of genomic DNA compared to control astrocytes. The extract of BM inhibited the formation of reactive species and DNA damage in a dose dependent manner. This data supports the traditional use of BM and indicates that this medicinal plant has a therapeutic potential in treatment or prevention of neurological diseases.


Subject(s)
Astrocytes/drug effects , Bacopa/chemistry , DNA Damage/drug effects , DNA/drug effects , Medicine, Ayurvedic , Nitric Oxide Donors/toxicity , Plant Extracts/therapeutic use , Reactive Oxygen Species/metabolism , S-Nitroso-N-Acetylpenicillamine/toxicity , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Comet Assay , Cytoprotection/drug effects , DNA Fragmentation/drug effects , L-Lactate Dehydrogenase , Rats , Rats, Wistar
11.
BMC Pharmacol ; 2: 18, 2002 Sep 13.
Article in English | MEDLINE | ID: mdl-12230634

ABSTRACT

BACKGROUND: S-nitrosoglutathione (GSNO) and S-nitroso-N-acetlypenicillamine (SNAP) are two of the most common sources of nitric oxide (NO) in the biomedical field. Vitamin C has been known to accelerate the decomposition of GSNO and SNAP increasing the release and availability of NO which is cytotoxic at non-physiological concentrations. The study investigates any potential detrimental effect of vitamin C and GSNO, vitamin C and SNAP on glucose metabolism in normotensive and normoglycemic dogs. RESULTS: The results showed that administration of vitamin C (50 mg/kg) and GSNO (35 mg/kg & 50 mg/kg), or vitamin C (50 mg/kg) and SNAP (10 mg/kg) to overnight fasted dogs resulted in significant elevation of the blood glucose levels, attaining maximum level at the 2.0 or 2.5 h time point postprandially. The elevated blood glucose levels were due to significant reduction in plasma insulin levels in the dogs treated with vitamin C and GSNO, or vitamin C and SNAP (P < 0.05). The decreased insulin response was associated with significant elevation of nitric oxide produced from GSNO and SNAP co-administered with vitamin C, as assessed by plasma nitrate/nitrite levels. CONCLUSIONS: The results indicate that enhanced NO release by vitamin C affects postprandial blood glucose and plasma insulin levels and the reduced glucose tolerance is mainly due to impaired insulin release. The clinical relevance of the findings of this study suggest that hypertensive diabetic patients treated with GSNO or SNAP, who are on vitamin C supplements may be more predisposed to further decrease in their glycemic control.


Subject(s)
Ascorbic Acid/pharmacology , Blood Glucose/drug effects , Hyperglycemia/chemically induced , S-Nitroso-N-Acetylpenicillamine/toxicity , S-Nitrosoglutathione/toxicity , Vasodilator Agents/toxicity , Animals , Dogs , Drug Synergism , Female , Male , Models, Animal
12.
FEBS Lett ; 510(3): 136-40, 2002 Jan 16.
Article in English | MEDLINE | ID: mdl-11801241

ABSTRACT

In the present study, we investigated the involvement of the mitochondrial permeability transition pore (PTP) in nitric oxide (NO)-induced plant cell death. NO donors such as sodium nitroprusside (SNP) and S-nitroso-N-acetylpenicillamine inhibited growth and caused death in suspension-cultured cells of Citrus sinensis. Cells treated with SNP showed chromatin condensation and fragmentation, characteristic of apoptosis. SNP caused loss of the mitochondrial membrane electrical potential, which was prevented by cyclosporin A (CsA), a specific inhibitor of PTP formation. CsA also prevented the nuclear apoptosis and subsequent Citrus cell death induced by NO. These findings indicate that mitochondrial PTP formation is involved in the signaling pathway by which NO induces apoptosis in cultured Citrus cells.


Subject(s)
Citrus/drug effects , Citrus/metabolism , Ion Channels , Membrane Proteins/metabolism , Nitric Oxide/toxicity , Apoptosis/drug effects , Cell Death/drug effects , Cell Division/drug effects , Cells, Cultured , Citrus/cytology , Cyclosporine/pharmacology , DNA Fragmentation , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Membrane Proteins/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Nitric Oxide Donors/toxicity , Nitroprusside/toxicity , S-Nitroso-N-Acetylpenicillamine/toxicity , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...