Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
J Inherit Metab Dis ; 44(2): 481-491, 2021 03.
Article in English | MEDLINE | ID: mdl-32882059

ABSTRACT

Sulfite oxidase (SO) deficiency is a disorder caused either by isolated deficiency of SO or by defects in the synthesis of its molybdenum cofactor. It is characterized biochemically by tissue sulfite accumulation. Patients present with seizures, progressive neurological damage, and basal ganglia abnormalities, the pathogenesis of which is not fully established. Treatment is supportive and largely ineffective. To address the pathophysiology of sulfite toxicity, we examined the effects of intrastriatal administration of sulfite in rats on antioxidant defenses, energy transfer, and mitogen-activated protein kinases (MAPK) and apoptosis pathways in rat striatum. Sulfite administration decreased glutathione (GSH) concentration and glutathione peroxidase, glucose-6-phosphate dehydrogenase, glutathione S-transferase, and glutathione reductase activities in striatal tissue. Creatine kinase (CK) activity, a crucial enzyme for cell energy transfer, was also decreased by sulfite. Superoxide dismutase-1 (SOD1) and catalase (CAT) proteins were increased, while heme oxygenase-1 (HO-1) was decreased. Additionally, sulfite altered phosphorylation of MAPK by decreasing of p38 and increasing of ERK. Sulfite further augmented the content of GSK-3ß, Bok, and cleaved caspase-3, indicating increased apoptosis. JP4-039 is a mitochondrial-targeted antioxidant that reaches higher intramitochondrial levels than other traditional antioxidants. Intraperitoneal injection of JP4-039 before sulfite administration preserved activity of antioxidant enzymes and CK. It also prevented or attenuated alterations in SOD1, CAT, and HO-1 protein content, as well as changes in p38, ERK, and apoptosis markers. In sum, oxidative stress and apoptosis induced by sulfite injection are prevented by JP4-039, identifying this molecule as a promising candidate for pharmacological treatment of SO-deficient patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/prevention & control , Antioxidants/pharmacology , Corpus Striatum/metabolism , Mitochondria/metabolism , Nitrogen Oxides/pharmacokinetics , Sulfite Oxidase/deficiency , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Catalase/metabolism , Cell Death/drug effects , Corpus Striatum/drug effects , Creatine Kinase/metabolism , Energy Transfer/drug effects , Glutathione Peroxidase/metabolism , Glutathione Peroxidase/pharmacology , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sulfites/metabolism , Superoxide Dismutase/metabolism
2.
FEBS Open Bio ; 11(1): 35-47, 2021 01.
Article in English | MEDLINE | ID: mdl-33179452

ABSTRACT

Cardiovascular disease has been associated with increased levels of reactive oxygen species (ROS). Recently, we have shown that a critical balance between cytosolic ROS and mitochondrial ROS is crucial in cardiovascular health and that modulation of mitochondrial ROS helps prevent detrimental effects of cytosolic ROS on endothelial cells (EC) in transgenic animals. Here, we report the development of a controlled delivery system for a mitochondria-targeted antioxidant, JP4-039, from an electrospun scaffold made of FDA-approved biocompatible polymeric nanofibers. We demonstrate that the active antioxidant moiety was preserved in released JP4-039 for over 72 h using electron paramagnetic resonance. We also show that both the initial burst release of the drug within the first 20 min and the ensuing slow and sustained release that occurred over the next 24 h improved tube formation in human coronary artery ECs (HCAEC) in vitro. Taken together, these findings suggest that electrospinning methods can be used to upload mitochondrial antioxidant (JP4-039) onto a biocompatible nanofibrous PLGA scaffold, and the uploaded drug (JP4-039) retains nitroxide antioxidant properties upon release from the scaffold, which in turn can reduce mitochondrial ROS and improve EC function in vitro.


Subject(s)
Antioxidants/administration & dosage , Drug Carriers/chemistry , Nanofibers/chemistry , Nitrogen Oxides/administration & dosage , Antioxidants/pharmacokinetics , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cell Line , Coronary Vessels/cytology , Coronary Vessels/pathology , Drug Liberation , Endothelial Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Nitrogen Oxides/pharmacokinetics , Reactive Oxygen Species/metabolism
3.
J Clin Pharmacol ; 59(5): 717-730, 2019 05.
Article in English | MEDLINE | ID: mdl-30703258

ABSTRACT

Nitroxyl (HNO) is a reactive nitrogen molecule that has potential therapeutic benefits for patients with acute heart failure. The results of the first-in-human study for BMS-986231, a novel HNO donor, are reported. The aim of this sequential cohort study was to evaluate the safety, tolerability, and pharmacokinetic profile of BMS-986231 after 24- and 48-hour intravenous infusions in healthy volunteers. Eighty subjects were randomized and dosed. Seven cohorts (stratum A) received BMS-986231 0.1, 0.33, 1, 3, 5, 10, and 15 µg/kg/min or placebo, infused over 24 hours. An additional cohort (stratum B) received 10 µg/kg/min or placebo, infused over 48 hours. Adverse events (AEs) were reported for 30 days after completion of infusion. Blood/urine samples were collected at regular intervals; other parameters (blood pressure, heart rate/rhythm, cardiac index) were also assessed. Headaches were the most commonly reported drug-related AE (48%) in those who received BMS-986231, although their severity was reduced by hydration. No other significant drug-related AEs were noted. BMS-986231 was associated with dose-dependent and well-tolerated reductions in systolic and diastolic blood pressure versus baseline; cardiac index, as measured noninvasively, was increased. BMS-986231 had no clinically significant effect on heart rate/rhythm or laboratory parameters. Its mean elimination half-life was 0.7-2.5 hours. BMS-986231 was safe and well-tolerated for up to 24 hours (15 µg/kg/min) or 48 hours (10 µg/kg/min), with a favorable hemodynamic profile observed. Ongoing studies continue to evaluate the potential benefit of BMS-986231 in patients with acute heart failure.


Subject(s)
Nitric Oxide Donors/pharmacokinetics , Nitrogen Oxides/pharmacokinetics , Adult , Blood Pressure/drug effects , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Heart Failure/drug therapy , Heart Rate/drug effects , Hemodynamics , Humans , Infusions, Intravenous , Male , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/blood , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/adverse effects , Nitrogen Oxides/blood , Nitrogen Oxides/pharmacology , Young Adult
4.
Nutr Health ; 24(3): 183-192, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30099933

ABSTRACT

BACKGROUND:: Ingestion of nitrate (NO3-)-containing vegetables, alcohol and polyphenols, separately, can reduce blood pressure (BP). However, the pharmacokinetic response to the combined ingestion of NO3- and polyphenol-rich or low polyphenol alcoholic beverages is unknown. AIM:: The aim of this study was to investigate how the consumption of low and high polyphenolic alcoholic beverages combined with a NO3--rich meal can influence NO3- metabolism and systemic BP. METHODS:: In a randomised, crossover trial, 12 normotensive males (age 25 ± 5 years) ingested an acute dose of NO3- (∼6.05 mmol) in the form of a green leafy salad, in combination with either a polyphenol-rich red wine (NIT-RW), a low polyphenol alcoholic beverage (vodka; NIT-A) or water (NIT-CON). Participants also consumed a low NO3- salad and water as a control (CON; ∼0.69 mmol NO3-). BP and plasma, salivary and urinary [NO3-] and nitrite ([NO2-]) were determined before and up to 5 h post ingestion. RESULTS:: Each NO3--rich condition elevated nitric oxide (NO) biomarkers when compared with CON ( P < 0.05). The peak rise in plasma [NO2-] occurred 1 h after NIT-RW (292 ± 210 nM) and 2 h after NIT-A (318 ± 186 nM) and NIT-CON (367 ± 179 nM). Systolic BP was reduced 2 h post consumption of NIT-RW (-4 mmHg), NIT-A (-3 mmHg) and NIT-CON (-2 mmHg) compared with CON ( P < 0.05). Diastolic BP and mean arterial pressure were also lower in NIT-RW and NIT-A compared with NIT-CON ( P < 0.05). CONCLUSIONS:: A NO3--rich meal, consumed with or without an alcoholic beverage, increases plasma [NO2-] and lowers systemic BP for 2-3 h post ingestion.


Subject(s)
Beverages , Blood Pressure/drug effects , Ethanol/pharmacology , Nitrates/pharmacology , Nitrogen Oxides/pharmacology , Polyphenols/pharmacology , Vegetables , Adult , Alcoholic Beverages , Cross-Over Studies , Female , Humans , Male , Meals , Nitrates/blood , Nitrates/pharmacokinetics , Nitric Oxide/blood , Nitrites/blood , Nitrogen Oxides/blood , Nitrogen Oxides/pharmacokinetics , Reference Values , Wine , Young Adult
5.
In Vivo ; 32(5): 1009-1023, 2018.
Article in English | MEDLINE | ID: mdl-30150422

ABSTRACT

BACKGROUND/AIM: The mitochondrial targeted GS-nitroxide, JP4-039, is an effective total body irradiation (TBI) mitigator when delivered intravenously (IV) up to 72 h after exposure. Effective systemic and localized administration to oral cavity/oropharynx and esophagus has been demonstrated. The objective of the study was to establish alternatives to IV administration suitable for JP4-039 delivery to mass casualties. MATERIALS AND METHODS: JP4-039 was administered to C57BL/6 mice by topically applied carboxy-methyl-cellulose microneedle arrays (MNAs) or by intramuscular (IM) injection. Three different formulations that have passed Food and Drug Administration review, namely Captisol, 2-hydroxypropyl-ß-cyclodextrin (cyclodextrin), and Miglyol-812-N, were used for drug delivery. Intraoral (IO) administration with each formulation was also evaluated. RESULTS: All tested formulations and MNAs successfully delivered JP4-039. However, IM delivery of the Miglyol-812-N displayed very efficient and highly reproducible radiation mitigation. CONCLUSION: Effective IM delivery of JP4-039 in animal models after TBI or partial-body irradiation suggested the use of the Miglyol-812-N formulation in both medical indications and radiation countermeasures.


Subject(s)
Drug Administration Routes , Drug Compounding , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/chemistry , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/chemistry , Administration, Intravenous , Administration, Oral , Animals , Apoptosis/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , Drug Stability , Female , Injections, Intramuscular , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Molecular Structure , Nitrogen Oxides/pharmacokinetics , Radiation Injuries, Experimental , Radiation, Ionizing , Radiation-Protective Agents/pharmacokinetics , Reproducibility of Results , Survival Rate , Whole-Body Irradiation
6.
Mol Pharmacol ; 93(5): 427-437, 2018 05.
Article in English | MEDLINE | ID: mdl-29476040

ABSTRACT

Glutathione-liganded binuclear dinitrosyl iron complex (glut-BDNIC) has been proposed to be a donor of nitric oxide (NO). This study was undertaken to investigate the mechanisms of vasoactivity, systemic hemodynamic effects, and pharmacokinetics of glut-BDNIC. To test the hypothesis that glut-BDNICs vasodilate by releasing NO in its reduced [nitroxyl (HNO)] state, a bioassay method of isolated, preconstricted ovine mesenteric arterial rings was used in the presence of selective scavengers of HNO or NO free radical (NO•); the vasodilatory effects of glut-BDNIC were found to have characteristics similar to those of an HNO donor and markedly different than an NO• donor. In addition, products of the reaction of glut-BDNIC with CPTIO [2-(4-carboxyphenyl)-4,4,5-tetramethyl imidazoline-1-oxyl-3-oxide] were found to have electron paramagnetic characteristics similar to those of an HNO donor compared with an NO• donor. In contrast to S-nitroso-glutathione, which was vasodilative both in vitro and in vivo, the potency of glut-BDNIC-mediated vasodilation was markedly diminished in both rats and sheep. Wire myography showed that plasma albumin contributed to this loss of hypotensive effects, an effect abolished by modification of the cysteine-thiol residue of albumin. High doses of glut-BDNIC caused long-lasting hypotension in rats that can be at least partially attributed to its long circulating half-life of ∼44 minutes. This study suggests that glut-BDNIC is an HNO donor, and that its vasoactive effects are modulated by binding to the cysteine residue of plasma proteins, such as albumin.


Subject(s)
Glutathione/metabolism , Hemodynamics/drug effects , Iron/metabolism , Iron/pharmacology , Nitrogen Oxides/metabolism , Nitrogen Oxides/pharmacology , Serum Albumin/metabolism , Animals , Antihypertensive Agents/pharmacology , Electron Spin Resonance Spectroscopy , Female , Iron/pharmacokinetics , Ligands , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Myography , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/pharmacokinetics , Rats , Sheep , Vasodilation/drug effects , Vasodilator Agents/pharmacology
7.
J Pharm Biomed Anal ; 150: 169-175, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-29245086

ABSTRACT

JP4-039 radio-protects prior to, and radio-mitigates after ionizing radiation by neutralizing reactive oxygen species. We developed and validated an LC-MS/MS assay for the quantitation of JP4-039 in murine plasma. Methanol protein precipitation of 50µL plasma was followed by isocratic reverse phase chromatography for a 6min run time, and electrospray positive mode ionization mass spectrometric detection. The plasma assay was linear from 1 to 1000ng/mL with appropriate accuracy (97.1-107.6%) and precision (3.7-12.5%CV), and fulfilled FDA guidance criteria. Recovery was 77.2-136.1% with moderate ionization enhancement (10.9-39.5%). Plasma freeze-thaw stability (98.8-104.2%), stability for 13.5 months at -80°C (93.1-105.6%), and stability for 4h at room temperature (94.2-97.6%) were all acceptable. Limited cross-validation to tissue homogenates suggested that these could also be analyzed for JP4-039 accurately. This assay has been directly applied to determine the pharmacokinetics of JP4-039 in C57BL/6 male mice after IV administration of 20mg/kg JP4-039 and will be extended to other studies of this agent.


Subject(s)
Chromatography, Reverse-Phase , Drug Monitoring/methods , Nitrogen Oxides/blood , Radiation-Protective Agents/metabolism , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Administration, Intravenous , Animals , Calibration , Chromatography, Reverse-Phase/standards , Cold Temperature , Drug Monitoring/standards , Drug Stability , Male , Mice, Inbred C57BL , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/pharmacokinetics , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacokinetics , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/standards , Tandem Mass Spectrometry/standards
8.
Bull Exp Biol Med ; 162(2): 207-210, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27913937

ABSTRACT

Subcutaneous injection of Oxacom with glutathione-bound dinitrosyl iron complex as the active principle produced a slower drop of mean BP and longer accumulation of protein-bound dinitrosyl iron complexes in whole blood and tissues than intravenous injection of this drug, while durations of hypotensive effect in both cases were practically identical. In contrast to intravenous injection of the drug, its subcutaneous administration was not characterized by a high concentration of protein-bound dinitrosyl iron complexes in the blood at the onset of experiment; in addition, accumulation of these NO forms in the lungs was more pronounced after subcutaneous injection than after intravenous one.


Subject(s)
Antihypertensive Agents/pharmacokinetics , Iron/pharmacokinetics , Nitric Oxide Donors/pharmacokinetics , Nitric Oxide/blood , Nitrogen Oxides/pharmacokinetics , Animals , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacology , Biotransformation , Injections, Intravenous , Injections, Subcutaneous , Iron/blood , Iron/pharmacology , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Muscle, Skeletal/metabolism , Myocardium/metabolism , Nitric Oxide Donors/blood , Nitric Oxide Donors/pharmacology , Nitrogen Oxides/blood , Nitrogen Oxides/pharmacology , Rats , Rats, Wistar
9.
Nitric Oxide ; 60: 1-9, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27565833

ABSTRACT

Nitrite infusion into the bloodstream has been shown to elicit vasodilation and protect against ischemia-reperfusion injury through nitric oxide (NO) release in hypoxic conditions. However, the mechanism by which nitrite-derived NO escapes scavenging by hemoglobin in the erythrocyte has not been fully elucidated, owing in part to the difficulty in measuring the reactions and transport on NO in vivo. We developed a mathematical model for an arteriole and surrounding tissue to examine the hypothesis that dinitrogen trioxide (N2O3) acts as a stable intermediate for preserving NO. Our simulations predict that with hypoxia and moderate nitrite concentrations, the N2O3 pathway can significantly preserve the NO produced by hemoglobin nitrite reductase in the erythrocyte and elevate NO reaching the smooth muscle cells. Nitrite retains its ability to increase NO bioavailability even at varying flow conditions, but there is minimal effect under normoxia or very low nitrite concentrations. Our model demonstrates a viable pathway for reconciling experimental findings of potentially beneficial effects of nitrite infusions despite previous models showing negligible NO elevation associated with hemoglobin nitrite reductase. Our results suggest that additional mechanisms may be needed to explain the efficacy of nitrite-induced vasodilation at low infusion concentrations.


Subject(s)
Arterioles/metabolism , Hypoxia/metabolism , Nitric Oxide/metabolism , Nitrites/pharmacology , Nitrogen Oxides/metabolism , Vasodilation/physiology , Animals , Arterioles/drug effects , Biological Availability , Blood Flow Velocity , Models, Biological , Nitrogen Oxides/pharmacokinetics , Oxygen/metabolism , Vasodilation/drug effects
10.
Redox Biol ; 8: 226-42, 2016 08.
Article in English | MEDLINE | ID: mdl-26827126

ABSTRACT

Free radicals, particularly reactive oxygen species (ROS), are involved in various pathologies, injuries related to radiation, ischemia-reperfusion or ageing. Unfortunately, it is virtually impossible to directly detect free radicals in vivo, but the redox status of the whole organism or particular organ can be studied in vivo by using magnetic resonance techniques (EPR and MRI) and paramagnetic stable free radicals - nitroxides. Here we review results obtained in vivo following the pharmacokinetics of nitroxides on experimental animals (and a few in humans) under various conditions. The focus was on conditions where the redox status has been altered by induced diseases or harmful agents, clearly demonstrating that various EPR/MRI/nitroxide combinations can reliably detect metabolically induced changes in the redox status of organs. These findings can improve our understanding of oxidative stress and provide a basis for studying the effectiveness of interventions aimed to modulate oxidative stress. Also, we anticipate that the in vivo EPR/MRI approach in studying the redox status can play a vital role in the clinical management of various pathologies in the years to come providing the development of adequate equipment and probes.


Subject(s)
Free Radicals/pharmacokinetics , Nitrogen Oxides/pharmacokinetics , Oxidative Stress , Reactive Oxygen Species/pharmacokinetics , Animals , Brain/metabolism , Brain/pathology , Electron Spin Resonance Spectroscopy , Free Radicals/isolation & purification , Humans , Magnetic Resonance Spectroscopy , Nitrogen Oxides/isolation & purification , Oxidation-Reduction , Reactive Oxygen Species/isolation & purification
11.
Magn Reson Med ; 76(3): 935-45, 2016 09.
Article in English | MEDLINE | ID: mdl-26414669

ABSTRACT

PURPOSE: The detailed in vivo T1 -weighted contrasting abilities of nitroxyl contrast agents, which have been used as redox responsive contrast agents in several magnetic resonance-based imaging modalities, in mouse brain were investigated. METHODS: Distribution and pharmacokinetics of five types of five-membered-ring nitroxyl radical compound were compared using T1 -weighted MRI. RESULTS: The blood-brain barrier (BBB) -impermeable 3-carboxy-2,2,5,5-tetramethylpyrrolidine-N-oxyl (CxP) could not be distributed in the brain. The slightly lipophilic 3-carbamoyl-2,2,5,5-tetramethylpyrrolidine-N-oxyl (CmP) showed slight distribution only in the ventricle, but not in the medulla and cortex. The amphiphilic 3-methoxy-carbonyl-2,2,5,5-tetramethyl-pyrrolidine-N-oxyl (MCP) had good initial uniform distribution in the brain and showed typical 2-phase signal decay profiles. A brain-seeking nitroxyl probe, acetoxymethyl-2,2,5,5-tetramethyl-pyrrolidine-N-oxyl-3-carboxylate (CxP-AM), showed an accumulating phase, and then its accumulation was maintained in the medulla and ventricle regions, but not in the cortex. The lipophilic 4-(N-methyl piperidine)-2,2,5,5-tetramethylpyrroline-N-oxyl (23c) was well distributed in the cortex and medulla, but slightly in the ventricle, and showed relatively rapid linear signal decay. CONCLUSION: Nitroxyl contrast agents equipped with a suitable lipophilic substitution group could be BBB-permeable functional contrast agents. MR redox imaging, which can estimate not only the redox characteristics but also the detailed distribution of the contrast agents, is a good candidate for a theranostic tool. Magn Reson Med 76:935-945, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Brain/metabolism , Contrast Media/pharmacokinetics , Magnetic Resonance Imaging/methods , Nitrogen Oxides/pharmacokinetics , Animals , Blood-Brain Barrier/diagnostic imaging , Capillary Permeability/physiology , Computer Simulation , Female , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Metabolic Clearance Rate , Mice , Mice, Inbred C3H , Models, Biological , Oxidation-Reduction , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
12.
Free Radic Res ; 49(8): 1038-47, 2015.
Article in English | MEDLINE | ID: mdl-25968953

ABSTRACT

Methamphetamine (METH)-induced neurotoxicity is associated with mitochondrial dysfunction and enhanced oxidative stress. The aims of the present study conducted in the mouse brain repetitively treated with METH were to (1) examine the redox status using the redox-sensitive imaging probe 3-methoxycarbonyl-2,2,5,5-tetramethylpiperidine-1-oxyl (MCP) and (2) non-invasively visualize the brain redox status with electron paramagnetic resonance (EPR) imaging. The rate of reduction of MCP was measured from a series of temporal EPR images of mouse heads, and this rate was used to construct a two-dimensional map of rate constants called a "redox map." The obtained redox map clearly illustrated the change in redox balance in the METH-treated mouse brain that is a known result of oxidative damage. Biochemical assays also showed that the level of thiobarbituric acid-reactive substance, an index of lipid peroxidation, was increased in mouse brains by METH. The enhanced reduction in MCP observed in mouse brains was remarkably suppressed by treatment with the dopamine synthase inhibitor, α-methyl-p-tyrosine, suggesting that enhancement of the reduction reaction of MCP resulted from enzymatic reduction in the mitochondrial respiratory chain. Furthermore, magnetic resonance imaging (MRI) of METH-treated mice using a blood-brain barrier (BBB)-impermeable paramagnetic contrast agent revealed BBB dysfunction after treatment with METH for 7 days. MRI also indicated that the impaired BBB recovered after withdrawal of METH. EPR imaging and MRI are useful tools not only for following changes in the redox status and BBB dysfunction in mouse brains repeatedly administered METH, but also for tracing the drug effect after withdrawal of METH.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System Stimulants/toxicity , Methamphetamine/toxicity , Oxidative Stress , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/drug effects , Brain/metabolism , Capillary Permeability/drug effects , Contrast Media/pharmacokinetics , Electron Spin Resonance Spectroscopy , Lipid Peroxidation , Locomotion/drug effects , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Nitrogen Oxides/pharmacokinetics , Organometallic Compounds/pharmacokinetics , Oxidation-Reduction
14.
Ter Arkh ; 86(9): 49-55, 2014.
Article in Russian | MEDLINE | ID: mdl-25518506

ABSTRACT

AIM: To examine the antihypertensive effect of the synthetic analogue of the endogenous nitric oxide donors in patients with grades 2-3 hypertension and uncomplicated hypertensive crisis (HC). SUBJECTS AND METHODS: The study included 30 male patients aged 35 to 73 years (mean age 55.5 ± 10.8 years). All the patients had grades 2-3 essential or secondary hypertension. Thirteen (43.3%) patients were observed to have signs of HC; 17 (56.7%) patients had persistent blood pressure (BP) elevation. A dinitrosyl iron complex was injected in a dose of 1.5 or 3 mg per kg of body weight. The purpose of its administration was to lower BP by at least 20% of its baseline level. RESULTS: No significant side effects associated with the administration of the test drug were recorded when the clinical trial protocol was implemented. All the patients reported fever and facial hyperemia during and 10-20 minutes after injection. They all (100%) showed efficient blood pressure reduction of at least 20% of the baseline level. Blood pressure changes were similar when the agent was administered in doses of 1.5 or 3 mg/kg. At 6-8 minutes after the drug was injected, there was a maximal decrease in blood pressure, then its gradual rise and stabilization at a lower level than the baseline one within the following 8 hours. There were no significant differences in the magnitude of a blood pressure reduction after administration of 1.5 and 3 mg/kg. CONCLUSION: The findings suggest that the dinitrosyl iron complex is highly effective in treating uncomplicated HC. The antihypertensive effect of the drug persists for 8 hours after its injection, which is very important during prehospital therapy. The drug is well tolerated by patients and causes an insignificant number of side effects.


Subject(s)
Blood Pressure/drug effects , Hemodynamics/drug effects , Hypertension , Iron , Nitrogen Oxides , Administration, Intravenous , Adult , Aged , Dose-Response Relationship, Drug , Drug Monitoring , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Iron/administration & dosage , Iron/adverse effects , Iron/pharmacokinetics , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/pharmacokinetics , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/adverse effects , Nitrogen Oxides/pharmacokinetics , Severity of Illness Index , Treatment Outcome
15.
In Vivo ; 28(4): 435-40, 2014.
Article in English | MEDLINE | ID: mdl-24982207

ABSTRACT

BACKGROUND/AIM: To determine whether Gramicidin S (GS)-nitroxide, JP4-039, esophageal radiation protection protected lung tumors in a transgenic model, LoxP-Stoop-LoxP Kristen Rat Sarcoma Viral oncogene (LSL-K-RAS) mice were administered intra-tracheal- Carbapenem-resistant Enterobacteriaceae (CRE) recombinase, bilateral lung tumors were confirmed at 11 weeks, then thoracic irradiation was delivered. MATERIALS AND METHODS: Mice received single-fraction 15 Gy or 24 Gy to both lungs, in subgroups receiving intraesophageal administration 10 min before irradiation of JP4-039 (in F15 emulsion) tumor size reduction and survival were investigated. Mice were followed for survival, and reduction in tumor size. RESULTS: There was no evidence of tumor radioprotection in mice receiving JP4-039/F15. CONCLUSION: Intraesophageal radioprotective small-molecule antioxidant therapy protects normal tissue but not tumor tissue in mice with transgenic lung tumors.


Subject(s)
Esophagus , Lung Neoplasms/radiotherapy , Nitrogen Oxides/administration & dosage , Organ Sparing Treatments , Radiation-Protective Agents/administration & dosage , Animals , Disease Models, Animal , Emulsions , Esophagus/drug effects , Esophagus/metabolism , Esophagus/radiation effects , Female , Genes, ras , Homologous Recombination , Integrases/genetics , Liposomes , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Nitrogen Oxides/pharmacokinetics , Radiation-Protective Agents/pharmacokinetics
16.
Clin Cancer Res ; 19(9): 2503-17, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23532887

ABSTRACT

PURPOSE: The study aimed to clarify the dynamics of tissue redox activity (TRA) in cancer progression and assess the importance of this parameter for therapeutic strategies. EXPERIMENTAL DESIGN: The experiments were carried out on brain tissues of neuroblastoma-bearing, glioma-bearing, and healthy mice. TRA was visualized in vivo by nitroxide-enhanced MRI on anesthetized animals or in vitro by electron paramagnetic resonance spectroscopy on isolated tissue specimens. Two biochemical parameters were analyzed in parallel: tissue total antioxidant capacity (TTAC) and plasma levels of matrix metalloproteinases (MMP). RESULTS: In the early stage of cancer, the brain tissues were characterized by a shorter-lived MRI signal than that from healthy brains (indicating a higher reducing activity for the nitroxide radical), which was accompanied by an enhancement of TTAC and MMP9 plasma levels. In the terminal stage of cancer, tissues in both hemispheres were characterized by a longer-lived MRI signal than in healthy brains (indicating a high-oxidative activity) that was accompanied by a decrease in TTAC and an increase in the MMP2/MMP9 plasma levels. Cancer progression also affected the redox potential of tissues distant from the primary tumor locus (liver and lung). Their oxidative status increased in both stages of cancer. CONCLUSIONS: The study shows that tissue redox balance is very sensitive to the progression of cancer and can be used as a diagnostic marker of carcinogenesis. The study also suggests that the noncancerous tissues of a cancer-bearing organism are susceptible to oxidative damage and should be considered a therapeutic target.


Subject(s)
Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Glioma/metabolism , Neuroblastoma/metabolism , Animals , Antioxidants/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Contrast Media/metabolism , Contrast Media/pharmacokinetics , Glioma/pathology , Half-Life , Humans , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neuroblastoma/pathology , Nitrogen Oxides/metabolism , Nitrogen Oxides/pharmacokinetics , Oxidation-Reduction
17.
Gastroenterology ; 143(4): 1027-36.e3, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22771506

ABSTRACT

BACKGROUND & AIMS: Drugs used to treat patients with ulcerative colitis are not always effective because of nonspecific distribution, metabolism in the gastrointestinal tract, and side effects. We designed a nitroxide radical-containing nanoparticle (RNP(O)) that accumulates specifically in the colon to suppress inflammation and reduce the undesirable side effects of nitroxide radicals. METHODS: RNP(O) was synthesized by assembly of an amphiphilic block copolymer that contains stable nitroxide radicals in an ether-linked hydrophobic side chain. Biodistribution of RNP(O) in mice was determined from radioisotope and electron spin resonance measurements. The effects of RNP(O) were determined in mice with dextran sodium sulfate (DSS)-induced colitis and compared with those of low-molecular-weight drugs (4-hydroxyl-2,2,6,6-tetramethylpiperidine-1-oxyl [TEMPOL] or mesalamine). RESULTS: RNP(O), with a diameter of 40 nm and a shell of poly(ethylene glycol), had a significantly greater level of accumulation in the colonic mucosa than low-molecular-weight TEMPOL or polystyrene latex particles. RNP(O) was not absorbed into the bloodstream through the intestinal wall, despite its long-term retention in the colon, which prevented its distribution to other parts of the body. Mice with DSS-induced colitis had significantly lower disease activity index and less inflammation following 7 days of oral administration of RNP(O) compared with mice with DSS-induced colitis or mice given low-molecular-weight TEMPOL or mesalamine. CONCLUSIONS: We designed an orally administered RNP(O) that accumulates specifically in the colons of mice with colitis and is more effective in reducing inflammation than low-molecular-weight TEMPOL or mesalamine. RNP(O) might be developed for treatment of patients with ulcerative colitis.


Subject(s)
Colitis/drug therapy , Colon/metabolism , Intestinal Mucosa/metabolism , Nanoparticles/therapeutic use , Nitrogen Oxides/pharmacokinetics , Nitrogen Oxides/therapeutic use , Analysis of Variance , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Colitis/chemically induced , Colitis/metabolism , Colon/enzymology , Cyclic N-Oxides/pharmacokinetics , Cyclic N-Oxides/therapeutic use , Dextran Sulfate , Interleukin-1beta/metabolism , Intestinal Mucosa/enzymology , Male , Mesalamine/therapeutic use , Mice , Mice, Inbred ICR , Nitrogen Oxides/blood , Peroxidase/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Severity of Illness Index , Spin Labels , Superoxides/metabolism , Survival Rate
18.
Biofizika ; 57(2): 331-7, 2012.
Article in Russian | MEDLINE | ID: mdl-22594291

ABSTRACT

Protein-bound dinitrosyl-iron complexes (DNIC) in rat whole blood and organs were studied after intravenous injection of this substance with glutathione ligand (DNIC-GH). The effect of DNIC-GH injection on NO level (including NO physiological forms) in hydrophobic areas of rat tissues was also studied in normal physiological blood circulation condition. It has been shown, that after DNIC-GH injection the concentration of protein-bound DNICs in rat whole blood and organs rapidly reached maximum values, and then gradually decreased, that pointed to decomposition of DNIC molecules, coupled with NO release. At the beginning of the experiment the rates of DNIC decay in rat heart and lung were substantially higher, as compared with those in liver and kidney. By spin trappping it has been demonstrated that DNIC-GH, as a source of NO physiological forms (including S-nitrosothiols), in normal physiological blood circulation influence heart more selectively, as compared with the other organs.


Subject(s)
Glutathione/pharmacokinetics , Iron/pharmacokinetics , Kidney/metabolism , Liver/metabolism , Myocardium/metabolism , Nitrogen Oxides/pharmacokinetics , Animals , Glutathione/pharmacology , Iron/pharmacology , Male , Nitric Oxide/metabolism , Nitrogen Oxides/pharmacology , Organ Specificity/drug effects , Rats , Rats, Wistar
19.
Biomaterials ; 33(9): 2723-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22209558

ABSTRACT

The development of improved, low toxicity, clinically viable nanomaterials that provide MRI contrast have tremendous potential to form the basis of translatable theranostic agents. Herein we describe a class of MRI visible materials based on lyotropic liquid crystal nanoparticles loaded with a paramagnetic nitroxide lipid. These readily synthesized nanoparticles achieved enhanced proton-relaxivities on the order of clinically used gadolinium complexes such as Omniscan™ without the use of heavy metal coordination complexes. Their low toxicity, high water solubility and colloidal stability in buffer resulted in them being well tolerated in vitro and in vivo. The nanoparticles were initially screened in vitro for cytotoxicity and subsequently a defined concentration range was tested in rats to determine the maximum tolerated dose. Pharmacokinetic profiles of the candidate nanoparticles were established in vivo on IV administration to rats. The lyotropic liquid crystal nanoparticles were proven to be effective liver MRI contrast agents. We have demonstrated the effective in vivo performance of a T1 enhancing, biocompatible, colloidally stable, amphiphilic MRI contrast agent that does not contain a metal.


Subject(s)
Fatty Alcohols , Liquid Crystals/chemistry , Magnetic Resonance Imaging/methods , Metals/chemistry , Nanoparticles , Nitrogen Oxides , Animals , CHO Cells , Cell Death/drug effects , Cricetinae , Cricetulus , Cryoelectron Microscopy , Fatty Alcohols/blood , Fatty Alcohols/chemistry , Fatty Alcohols/pharmacokinetics , HEK293 Cells , Humans , Liquid Crystals/toxicity , Male , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Nitrogen Oxides/blood , Nitrogen Oxides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Scattering, Small Angle , Synchrotrons , X-Ray Diffraction
20.
Kardiologiia ; 51(11): 28-37, 2011.
Article in Russian | MEDLINE | ID: mdl-22117768

ABSTRACT

On the basis of earlier executed studies of hypotensive effect of dinitrosyl iron complexes (DNIC) with glutathione, the drug has been created in industrial conditions named oxacom. Preliminary pharmacological studies of oxacom have not revealed negative qualities. The drug has been now tested in 14 healthy men in whom at single intravenous introduction it caused typical response - a decrease of diastolic as well as systolic arterial pressure on 24-27 mmHg through 3-4 min with subsequent very slow restoration in 8-10 hours. The heart rate after initial rise was quickly normalized. Echocardiography revealed unaltered cardiac output in spite of reduced cardiac filling by 28%. The multilateral analysis of clinical and biochemical data has revealed an absence of essential alterations which could lead to pathological consequences. The drug is recommended for carrying out of the second phase of clinical trial. The comparative study of the efficiency of hypotensive action of oxacom, S-nitrosoglutathione (GS-NO) and sodium nitrite (NO2) in rats has shown that the duration of effect was the greatest at oxacom action.


Subject(s)
Blood Pressure/drug effects , Glutathione , Hypertension/drug therapy , Iron , Nitrogen Oxides , S-Nitrosoglutathione/pharmacokinetics , Sodium Nitrite/pharmacokinetics , Adult , Animals , Biological Availability , Drug Evaluation, Preclinical/methods , Drug Monitoring/methods , Glutathione/administration & dosage , Glutathione/adverse effects , Glutathione/pharmacokinetics , Glutathione/pharmacology , Humans , Hypertension/metabolism , Hypertension/physiopathology , Hypotension/chemically induced , Infusions, Intravenous , Iron/administration & dosage , Iron/adverse effects , Iron/pharmacokinetics , Iron/pharmacology , Male , Nitric Oxide/metabolism , Nitrogen Oxides/administration & dosage , Nitrogen Oxides/adverse effects , Nitrogen Oxides/pharmacokinetics , Nitrogen Oxides/pharmacology , Rats , Rats, Wistar , Therapeutic Equivalency , Therapies, Investigational , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...