Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 171
1.
Eur J Pharm Sci ; 131: 159-166, 2019 Apr 01.
Article En | MEDLINE | ID: mdl-30779982

Ischemic heart conditions are among the main causes of sudden cardiac death worldwide. One of the strategies for avoiding myocardial infarction is the low-dose, prophylactic use of acetylsalicylic acid (ASA), an inhibitor of platelet aggregation. To avoid the gastrointestinal damage, ASA prodrugs bearing nitric oxide (NO)-donating moiety covalently conjugated to ASA have been synthesized and evaluated extensively worldwide. Herein the synthesis of a new hybrid ASA ester covalently attached to the NO donor linsidomine, an active metabolite of molsidomine (MOL) is reported. Cell viability assay and hemolysis tests were performed in H9c2 cells and rat erythrocytes, respectively. Our new compound, the ERJ-500 not affected negatively the viability of living cells in the concentration range of 100 nM to 100 µM. Using the ex vivo Langendorff method on hearts originated from female rats, compound ERJ-500 displayed a dose-dependent, outwashable vasodilative effect in coronary arteries. Vasodilation was observed on isolated working heart model as well, with elevated stroke volume in hearts treated with ERJ-500. Furthermore, a decreased infarct size was also noticed in ERJ-500 treated hearts after ischemia/reperfusion. Based on these observations it can be expected that our new hybrid ASA may contribute to new pharmacological tool in the therapy of ischemic heart conditions and associated syndromes.


Aspirin/analogs & derivatives , Aspirin/administration & dosage , Heart/drug effects , Molsidomine/administration & dosage , Nitric Oxide/administration & dosage , Vasodilator Agents/administration & dosage , Animals , Aspirin/pharmacology , Cell Line , Coronary Circulation/drug effects , Erythrocytes/drug effects , Female , Heart/physiology , Heart Rate/drug effects , Hemolysis/drug effects , Rats, Sprague-Dawley , Vasodilation/drug effects
2.
J Psychopharmacol ; 32(6): 711-722, 2018 06.
Article En | MEDLINE | ID: mdl-29737230

BACKGROUND: An excitatory imbalance in the hypothalamus of rodents caused by local chemical stimulation elicits fear-related defensive reactions such as escape and freezing. In addition, these panic attack-like defensive reactions induced by hypothalamic neurons may cause antinociception. However, there is a shortage of studies showing the participation of the anterior hypothalamic nucleus in these adaptive defensive mechanisms. Nitric oxide (NO) donors have been shown to evoke fear-related defensive responses when microinjected into paralimbic and limbic structures, and this excitatory neuromodulation can recruit the glutamatergic system. AIMS: The aim of this work was to investigate the influence of the glutamatergic system in the nitrergic effects on fear-related defensive responses organised by anterior hypothalamic neurons. METHODS: The present study evaluates the effects of the molsidomine active metabolite SIN-1 NO donor administered into the anterior hypothalamus (AH) of mice at different concentrations (75, 150 and 300 nmol/0.1 µL). Then, we investigated the effects of pre-treatment of the AH with AP-7 (an N-methyl-d-aspartate (NMDA) receptor-selective antagonist; 0.02, 0.2 and 2 nmol/0.1 µL) on the behavioural and antinociceptive effects provoked by AH chemical stimulation with SIN-1 microinjections. RESULTS: The 300 nmol dose of SIN-1 was the most effective at causing panic-like defensive behaviours followed by a significant antinociceptive response. In addition, both of these effects were attenuated or inhibited by AH pre-treatment with AP-7. CONCLUSIONS: These findings suggest that the panicogenic and antinociceptive effects evoked by intra-AH microinjections of SIN-1 depend on NMDA receptor activation.


Fear/drug effects , Molsidomine/analogs & derivatives , Nitric Oxide Donors/administration & dosage , Panic/drug effects , 2-Amino-5-phosphonovalerate/administration & dosage , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Hypothalamus, Anterior/drug effects , Hypothalamus, Anterior/metabolism , Male , Mice , Mice, Inbred C57BL , Microinjections , Molsidomine/administration & dosage , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Biotech Histochem ; 92(1): 68-77, 2017.
Article En | MEDLINE | ID: mdl-28166419

We investigated the protective and therapeutic effects of molsidomine (MOL) in a rat model of whole brain radiotherapy (RT). Forty female rats were divided into five groups of eight: group 1, control; group 2, 15 Gy single dose RT (RT); group 3, 4 mg/kg MOL treated for 5 days (MOL); group 4, 4 mg/kg MOL for 5 days, 10 days after RT treatment (RT + MOL); group 5, 4 mg/kg MOL treatment for 5 days before RT treatment and for 5 days after RT treatment (MOL + RT). All rats were sacrificed on day 16. Neurodegenerative changes in the brain and tissue levels of oxidants and antioxidants were evaluated. The oxidative parameters were increased and antioxidant status was decreased in group RT compared to groups MOL + RT and RT + MOL. Histopathological examination showed that treatment with MOL after RT application and treatment with MOL before RT treatment decreased neuronal degeneration. No difference in neuronal appearance was found between groups RT + MOL and MOL + RT. MOL treatment protected the nervous system of rats and may be a treatment option for preventing RT induced neural injury.


Brain/radiation effects , Molsidomine/therapeutic use , Radiation Injuries, Experimental/prevention & control , Animals , Brain/metabolism , Female , Glutathione , Malondialdehyde , Molsidomine/administration & dosage , Radiation, Ionizing , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/therapeutic use , Rats , Superoxide Dismutase
4.
Redox Biol ; 11: 438-455, 2017 04.
Article En | MEDLINE | ID: mdl-28086193

Reactive oxygen and nitrogen species (ROS/RNS) play an important role in the regulation of cardiac function. Increase in ROS/RNS concentration results in lipid and protein oxidation and is often associated with onset and/or progression of many cardiovascular disorders. However, interplay between lipid and protein modifications has not been simultaneously studied in detail so far. Biomolecule carbonylation is one of the most common biomarkers of oxidative stress. Using a dynamic model of nitroxidative stress we demonstrated rapid changes in biomolecule carbonylation in rat cardiomyocytes. Levels of carbonylated species increased as early as 15min upon treatment with the peroxynitrite donor, 3-morpholinosydnonimine (SIN-1), and decreased to values close to control after 16h. Total (lipids+proteins) vs. protein-specific carbonylation showed different dynamics, with a significant increase in protein-bound carbonyls at later time points. Treatment with SIN-1 in combination with inhibitors of proteasomal and autophagy/lysosomal degradation pathways allowed confirmation of a significant role of the proteasome in the degradation of carbonylated proteins, whereas lipid carbonylation increased in the presence of autophagy/lysosomal inhibitors. Electrophilic aldehydes and ketones formed by lipid peroxidation were identified and relatively quantified using LC-MS/MS. Molecular identity of reactive species was used for data-driven analysis of their protein targets. Combination of different enrichment strategies with LC-MS/MS analysis allowed identification of more than 167 unique proteins with 332 sites modified by electrophilic lipid peroxidation products. Gene ontology analysis of modified proteins demonstrated enrichment of several functional categories including proteins involved in cytoskeleton, extracellular matrix, ion channels and their regulation. Using calcium mobilization assays, the effect of nitroxidative stress on the activity of several ion channels was further confirmed.


Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Protein Carbonylation/genetics , Reactive Nitrogen Species/metabolism , Aldehydes/metabolism , Animals , Autophagy/genetics , Ketones/metabolism , Lipid Peroxidation/genetics , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Myocytes, Cardiac/drug effects , Nitrogen/metabolism , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Rats , Reactive Oxygen Species/metabolism
5.
Pharmacol Rep ; 69(1): 29-35, 2017 Feb.
Article En | MEDLINE | ID: mdl-27764702

BACKGROUND: Interaction between dopaminergic and nitrergic neurotransmission in the brain plays a crucial role in the control of motor function and in the regulation of blood pressure (BP). In Parkinson's disease (PD), dopaminergic denervation of the striatum leads to disturbances in the nitrergic system in the basal ganglia. Recently, it has been demonstrated that addition of a low dose of the nitric oxide donor molsidomine to l-DOPA therapy improves dopaminergic neurotransmission in the denervated nigrostriatal system and weakens dyskinesias in rodent models of the disease. METHODS: The aim of the present study was to examine the impact of chronic administration of molsidomine (2mg/kg) and l-DOPA (25mg/kg), alone and in combination, on systolic (SBP) and diastolic (DBP) blood pressure in the anesthetized, unilaterally 6-OHDA-lesioned rats. The measurement of SBP and DBP was performed 24h after the penultimate and immediately after the last drug doses. RESULTS: In 6-OHDA-lesioned rats receiving saline, spontaneous, small decreases in SBP and DBP were observed during the measurements lasting 60min. Administration of molsidomine alone or in combination with l-DOPA distinctly decreased the BP in 6-OHDA-lesioned rats already after 10min compared to those treated with saline or l-DOPA alone, respectively. In both groups, the molsidomine-mediated declines in BP persisted till the end of measurement but they disappeared after 24h. CONCLUSIONS: Our results indicate that in this PD model molsidomine evokes a short-lasting decrease in BP in contrast to conventional antihypertensive drugs that maintain long-term effect and worsen orthostatic hypotension in parkinsonian patients.


Antiparkinson Agents/administration & dosage , Blood Pressure/drug effects , Levodopa/administration & dosage , Molsidomine/administration & dosage , Nitric Oxide Donors/administration & dosage , Oxidopamine/toxicity , Animals , Blood Pressure/physiology , Male , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Rats , Rats, Wistar
6.
PLoS One ; 11(10): e0164893, 2016.
Article En | MEDLINE | ID: mdl-27760216

Embryonic VE-Cadherin-expressing progenitors (eVE-Cad+), including hemogenic endothelium, have been shown to generate hematopoietic stem cells and a variety of other progenitors, including mesoangioblasts, or MABs. MABs are vessel-associated progenitors with multilineage mesodermal differentiation potential that can physiologically contribute to skeletal muscle development and regeneration, and have been used in an ex vivo cell therapy setting for the treatment of muscular dystrophy. There is currently a therapeutic need for molecules that could improve the efficacy of cell therapy protocols; one such good candidate is nitric oxide. Several studies in animal models of muscle dystrophy have demonstrated that nitric oxide donors provide several beneficial effects, including modulation of the activity of endogenous cell populations involved in muscle repair and the delay of muscle degeneration. Here we used a genetic lineage tracing approach to investigate whether the therapeutic effect of nitric oxide in muscle repair could derive from an improvement in the myogenic differentiation of eVE-Cad+ progenitors during embryogenesis. We show that early in vivo treatment with the nitric oxide donor molsidomine enhances eVE-Cad+ contribution to embryonic and fetal myogenesis, and that this effect could originate from a modulation of the properties of yolk sac hemogenic endothelium.


Hemangioblasts/cytology , Molsidomine/administration & dosage , Muscle Development/drug effects , Nitric Oxide Donors/administration & dosage , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Differentiation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Hemangioblasts/drug effects , Hemangioblasts/metabolism , Mice , Molsidomine/pharmacology , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Animal/pathology , Nitric Oxide Donors/pharmacology
7.
J Pharm Sci ; 105(6): 1952-1959, 2016 06.
Article En | MEDLINE | ID: mdl-27238492

Composites of cardiovascular drug molsidomine with silica materials (unmodified and mercaptopropyl modified) were prepared by 2 methods, adsorption and sol-gel technology. The effects of sol pH and release medium pH (1.6 and 7.4) as well as molsidomine loading on the drug release kinetics were also investigated. Mechanisms of molsidomine release from all the synthesized composites were elucidated. The obtained results showed that different principles of the composites formation (adsorption or sol-gel) lead to their different release behavior because the composites obtained by the indicated methods differ by distribution of the drug over the silica matrixes and their capability to degradation. The drug release from the composites prepared by adsorption is characterized by a high burst effect, sustained release up to 36 h irrespective of release medium pH. The release behavior of sol-gel composites depends on the amount of the loaded drug and release medium pH. These effects were explained by different stability of the sol-gel composites with high and low loading in acidic and neutral media. In general case, the ascertained effects are independent on chemistry of the silica surface organic groups.


Cardiovascular Agents/pharmacokinetics , Drug Delivery Systems/trends , Drug Liberation , Hydrophobic and Hydrophilic Interactions , Molsidomine/pharmacokinetics , Silicon Dioxide/pharmacokinetics , Cardiovascular Agents/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Drug Delivery Systems/methods , Drug Liberation/physiology , Molsidomine/administration & dosage , Silicon Dioxide/administration & dosage , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacokinetics
8.
World Neurosurg ; 91: 673.e11-8, 2016 Jul.
Article En | MEDLINE | ID: mdl-27109628

BACKGROUND: A case of hyperacute vasospasm, indicating a poor prognosis after aneurysmal subarachnoid hemorrhage (SAH), is reported, and a review is presented of the literature addressing use of nitric oxide (NO) donors in cases of refractory vasospasm and recurrent delayed cortical ischemias (DCI). CASE DESCRIPTION: A 65-year-old woman was admitted within 1 hour after aneurysmal SAH (Hunt and Hess grade III, Fisher modified by Frontera grade IV). A hyperacute vasospasm had been confirmed arteriographically, the right middle cerebral artery (MCA) aneurysm was immediately coiled and a standard antivasospastic therapy was started. Within 48 hours, the patient developed cerebral vasospasm with DCI. Because the standard therapy failed to control clinical symptoms and to address severe vasospasm, an individualized rescue treatment with NO donors was initiated. A continuous intravenous molsidomine infusion was started and clinical stabilization was achieved for a week (Hunt and Hess grade I; World Federation of Neurological Surgeons grade I; Glasgow Coma Scale score, 15) after which vasospasm and DCI recurred. During a subsequent DCI, we escalated NO donor therapy by adding intraventricular boluses of sodium nitroprusside (SNP). Over the course of the following 22 days, 7 transient DCIs (Glasgow Coma Scale score, 8) were treated with boluses of SNP during continued molsidomine therapy and each time vasospasm and DCI were completely reversed. Despite initial poor prognosis, the clinical outcome was excellent; at 3, 6, and 12 months follow-up the patient's modified National Institutes of Health-Stroke Scale and modified Rankin Scale scores were 0, with no cognitive deficits. CONCLUSIONS: The review of the literature suggested that combined intravenous molsidomine with intraventricular SNP treatment reversed refractory, recurrent vasospasm and DCIs probably by addressing the hemoglobin NO sink effect, NO depletion, and decreased NO availability after aneurysmal SAH.


Nitric Oxide/administration & dosage , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/drug therapy , Aged , Female , Glasgow Coma Scale , Glasgow Outcome Scale , Humans , Injections, Intravenous/methods , Injections, Intraventricular/methods , Molsidomine/administration & dosage , Nitroprusside/administration & dosage , Treatment Outcome
9.
Behav Neurosci ; 129(1): 50-61, 2015 Feb.
Article En | MEDLINE | ID: mdl-25621792

Nitric oxide (NO) is an important molecule for the proper development and function of the central nervous system. In this study, we investigated the behavioral alterations in the neuronal NO synthase knockout mice (NOS1 KO) with a deficient NO production mechanism in the brain, characterizing it as a potential rodent model for attention deficit hyperactivity disorder (ADHD). NOS1 KO exhibited higher locomotor activity than their wildtype counterparts in a novel environment, as measured by open field (OF) test. In a 2-way active avoidance paradigm (TWAA), we found sex-dependent effects, where male KO displayed deficits in avoidance and escape behavior, sustained higher incidences of shuttle crossings, and higher incidences of intertrial interval crossings, suggesting learning, and/or performance impairments. On the other hand, female KO demonstrated few deficits in TWAA. Molsidomine (MSD), a NO donor, rescued TWAA deficits in male KO when acutely administered before training. In a passive avoidance paradigm, KO of both sexes displayed significantly shorter step-through latencies after training. Further, abnormal spontaneous motor activity rhythms were found in the KO during the dark phase of the day, indicating dysregulation of rhythmic activities. These data indicate that NOS1 KO mimics certain ADHD-like behaviors and could potentially serve as a novel rodent model for ADHD.


Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/psychology , Disease Models, Animal , Nitric Oxide Synthase Type I/physiology , Animals , Attention Deficit Disorder with Hyperactivity/prevention & control , Avoidance Learning/drug effects , Avoidance Learning/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molsidomine/administration & dosage , Motor Activity/drug effects , Nitric Oxide Synthase Type I/genetics
10.
Neurobiol Dis ; 73: 49-59, 2015 Jan.
Article En | MEDLINE | ID: mdl-25281315

Nitric oxide (NO), a gaseous messenger molecule synthesized by nitric oxide synthase (NOS), plays a pivotal role in integrating dopamine transmission in the basal ganglia and has been implicated in the pathogenesis of Parkinson disease (PD). To study the role of the nitrergic system in l-DOPA-induced dyskinesia (LID), we assessed the effect of the pharmacological manipulation of NO levels and NO/cyclic guanosine monophosphate (cGMP) signaling on LID in the Pitx3(-/-) aphakia mouse, a genetic model of PD. To evaluate the effect of decreased NO signaling on the development of LID, Pitx3(-/-) mice were chronically treated with l-DOPA and 7-nitroindazole (7-NI, a neuronal NOS inhibitor). To evaluate its effect on the expression of established LID, 7-NI was administered acutely to dyskinetic mice. The chronic 7-NI treatment attenuated the development of LID in the Pitx3(-/-) mice, and the sub-acute 7-NI treatment attenuated established dyskinesia without affecting the beneficial therapeutic effect of l-DOPA. Moreover, 7-NI significantly reduced FosB and pAcH3 expression in the acutely and chronically l-DOPA-treated mice. We also examined how increasing NO/cGMP signaling affects LID expression by acutely administering molsidomine (an NO donor) or zaprinast (a cGMP phosphodiesterase 5-PDE5 inhibitor) before l-DOPA in mice with established dyskinesia. Paradoxically, the administration of either of these drugs also significantly diminished the expression of established LID; however, the effect occurred at the expense of the antiparkinsonian l-DOPA properties. We demonstrate that targeting the NO/cGMP signaling pathway reduces dyskinetic behaviors and molecular markers, but only the 7-NI treatment preserved the antiparkinsonian effect of l-DOPA, indicating that NOS inhibitors represent a potential therapy to reduce LID.


Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/drug therapy , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Indazoles/therapeutic use , Transcription Factors/deficiency , Animals , Antiparkinson Agents/toxicity , Benserazide/therapeutic use , Cell Count , Corpus Striatum/drug effects , Disease Models, Animal , Dopamine Agents/toxicity , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/pathology , Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Levodopa/toxicity , Mice , Mice, Knockout , Molsidomine/administration & dosage , Nitric Oxide Donors/administration & dosage , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Transcription Factors/genetics
11.
Adv Ther ; 31(1): 91-106, 2014 Jan.
Article En | MEDLINE | ID: mdl-24307221

INTRODUCTION: The direct nitric oxide donor molsidomine is commonly used to relieve symptoms in chronic angina thanks to its vasodilatory properties that induce both a reduction in myocardial oxygen demand and an increase in coronary blood flow. The objective of this study was to compare the short-term effect of molsidomine 16 mg once daily (Coruno(®), Therabel Pharmaceuticals Ltd, Dublin, Ireland) in patients with stable angina previously on molsidomine 8 mg twice daily, in care of general practitioners (GPs) versus cardiologists. METHODS: A total of 53 and 80 patients treated by GPs and cardiologists, respectively, took part in this multicenter, open-label clinical trial. Frequency of anginal attacks, short-acting nitroderivative tablets consumption, and subjective clinical status were evaluated, as was tolerability to molsidomine through the reporting of adverse events (AEs), the recording of vital parameters-resting blood pressure, heart rate, and electrocardiogram-and routine blood analyses. RESULTS: Although demographic and clinical characteristics were significantly different in GPs' compared with cardiologists' patients, the effect of either the 8 mg or the 16 mg molsidomine formulation on anginal crises and nitroderivatives consumption was similar in both patient cohorts, with a trend for the 16 mg formulation to be more efficient on symptoms in elderly patients. Subjective assessment of the beneficial effect of molsidomine 16 versus 8 mg was comparable in GPs' and cardiologists' patients independently of age, "no change" being the most often reported item. Self-evaluation of functional capacity in elderly showed on the whole no difference between the two patient cohorts, only magnitude of pace and total score on molsidomine 16 mg being higher in cardiologists' compared with GPs' patients, and total score in cardiologists' patients higher on molsidomine 16 mg compared with 8 mg. Incidences of AEs and drug-related AEs, as well as proportions of patients reporting such AEs, were similar between GPs' and cardiologists' patient cohorts as between molsidomine 8 and 16 mg formulations. Molsidomine 16 mg once daily had no clinically significant effect on blood pressure, heart rate, electrocardiogram or blood parameters, and compliance with treatment was excellent in whatever patient cohort. CONCLUSION: Despite significant demographic and clinical differences between patients in care of GPs and cardiologists, molsidomine was equally efficient in these two patient cohorts, with a trend towards less anginal attacks in the elderly when treated with the 16 mg compared with the 8 mg formulation. Compliance and tolerability to the drug were excellent in both patient cohorts.


Angina, Stable/drug therapy , Molsidomine/administration & dosage , Vasodilator Agents/administration & dosage , Aged , Aged, 80 and over , Cardiology , Drug Administration Schedule , Female , General Practice , Humans , Male , Middle Aged , Treatment Outcome
12.
Arch Med Res ; 44(7): 521-8, 2013 Oct.
Article En | MEDLINE | ID: mdl-24120390

BACKGROUND AND AIMS: Despite its beneficial effects, cisplatin has considerable nephrotoxic, ototoxic, neurotoxic and hepatotoxic side effects. It has been documented that reactive oxygen radical species are involved with the pathophysiology of cisplatin-induced hepatotoxicity. Molsidomine (MOL) can exert antioxidant and anti-inflammatory effects. Therefore, the current study was planned to determine the effects of cisplatin on the liver oxidant/antioxidant system and the possible protective effects of (MOL) on liver toxicity. METHODS: Animals were divided into four groups as follows: (1) control; (2) MOL; (3) cisplatin and (4) MOL plus cisplatin group. Biochemical and histopathological evaluations were performed on the extracted liver tissue. Also, serum levels of serum aspartate transaminase (AST) and serum alanine transaminase (ALT) were determined. RESULTS: Our results clearly indicated that liver antioxidant enzyme activities and ALT levels were significantly decreased, whereas lipid peroxidation and neutrophil accumulation were increased in the cisplatin-treated animals (5 mg/kg single dose, i.p.) compared to the control rats. MOL treatment (4 mg/kg/day, i.p.) for 3 consecutive days provided a significant protection against cisplatin-induced hazardous changes in the liver tissue. Our histopathological findings including caspase-3 activity were also in accordance with the biochemical results. CONCLUSIONS: We propose that MOL acts in the liver as a potent scavenger of free radicals, anti-inflammatory and anti-apoptotic effects to prevent the toxic effects of cisplatin, both at the biochemical and histopathological levels.


Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Free Radical Scavengers/administration & dosage , Hepatocytes/drug effects , Liver/drug effects , Molsidomine/administration & dosage , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Caspase 3/metabolism , Cisplatin/therapeutic use , Hepatocytes/enzymology , Hepatocytes/pathology , Lipid Peroxidation , Liver/enzymology , Liver/pathology , Male , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology
13.
Eur J Pharmacol ; 679(1-3): 40-50, 2012 Mar 15.
Article En | MEDLINE | ID: mdl-22293370

We previously reported that both nitric oxide (NO) generated from NO synthase by bombesin and NO generated from SIN-1 (NO donor) activate the brain cyclooxygenase (COX) (COX-1 for bombesin), thereby eliciting the secretion of both catecholamines (CA) from the adrenal medulla by brain thromboxane A(2)-mediated mechanisms in rats. NO exerts its effects via not only soluble guanylate cyclase, but also protein S-nitrosylation, covalent modification of a protein cysteine thiol. In this study, we clarified the central mechanisms involved in the bombesin-induced elevation of plasma CA with regard to the relationship between NO and COX-1 using anesthetized rats. Bombesin (1 nmol/animal, i.c.v.)-induced elevation of plasma CA was attenuated by carboxy-PTIO (NO scavenger) (0.5 and 2.5 µmol/animal, i.c.v.), but was not influenced by ODQ (soluble guanylate cyclase inhibitor) (100 and 300 nmol/animal, i.c.v.). The bombesin-induced response was effectively reduced by dithiothreitol (thiol-reducing reagent) (0.4 and 1.9 µmol/kg/animal, i.c.v.) and by N-ethylmaleimide (thiol-alkylating reagent) (0.5 and 2.4 µmol/kg/animal, i.c.v.). The doses of dithiothreitol also reduced the SIN-1 (1.2 µmol/animal, i.c.v.)-induced elevation of plasma CA, but had no effect on the U-46619 (thromboxane A(2) analog) (100 nmol/animal, i.c.v.)-induced elevation of plasma CA even at higher doses (1.9 and 9.7 µmol/kg/animal, i.c.v.). Immunohistochemical studies demonstrated that the bombesin increased S-nitroso-cysteine-positive cells co-localized with COX-1 in the spinally projecting neurons of the hypothalamic paraventricular nucleus (PVN). Taken together, endogenous NO seems to mediate centrally administered bombesin-induced activation of adrenomedullary outflow at least in part by S-nitrosylation of COX-1 in the spinally projecting PVN neurons in rats.


Adrenal Medulla/drug effects , Bombesin/pharmacology , Catecholamines/blood , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/administration & dosage , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adrenal Medulla/metabolism , Animals , Benzoates/administration & dosage , Benzoates/pharmacology , Bombesin/administration & dosage , Bombesin/antagonists & inhibitors , Brain/drug effects , Cysteine/analogs & derivatives , Cysteine/metabolism , Dithiothreitol/administration & dosage , Dithiothreitol/pharmacology , Dose-Response Relationship, Drug , Ethylmaleimide/administration & dosage , Ethylmaleimide/pharmacology , Imidazoles/administration & dosage , Imidazoles/pharmacology , Injections, Intraventricular , Male , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Molsidomine/antagonists & inhibitors , Molsidomine/pharmacology , Nitric Oxide Donors/administration & dosage , Nitric Oxide Donors/pharmacology , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Rats , Rats, Wistar , S-Nitrosothiols/metabolism , Sulfhydryl Reagents/administration & dosage , Sulfhydryl Reagents/pharmacology
14.
J Neuroinflammation ; 8: 78, 2011 Jul 06.
Article En | MEDLINE | ID: mdl-21733162

BACKGROUND: Traumatic brain injury (TBI) induces primary and secondary damage in both the endothelium and the brain parenchyma, collectively termed the neurovascular unit. While neurons die quickly by necrosis, a vicious cycle of secondary injury in endothelial cells exacerbates the initial injury in the neurovascular unit following TBI. In activated endothelial cells, excessive superoxide reacts with nitric oxide (NO) to form peroxynitrite. Peroxynitrite has been implicated in blood brain barrier (BBB) leakage, altered metabolic function, and neurobehavioral impairment. S-nitrosoglutathione (GSNO), a nitrosylation-based signaling molecule, was reported not only to reduce brain levels of peroxynitrite and oxidative metabolites but also to improve neurological function in TBI, stroke, and spinal cord injury. Therefore, we investigated whether GSNO promotes the neurorepair process by reducing the levels of peroxynitrite and the degree of oxidative injury. METHODS: TBI was induced by controlled cortical impact (CCI) in adult male rats. GSNO or 3-Morpholino-sydnonimine (SIN-1) (50 µg/kg body weight) was administered orally two hours following CCI. The same dose was repeated daily until endpoints. GSNO-treated (GSNO group) or SIN-1-treated (SIN-1 group) injured animals were compared with vehicle-treated injured animals (TBI group) and vehicle-treated sham-operated animals (Sham group) in terms of peroxynitrite, NO, glutathione (GSH), lipid peroxidation, blood brain barrier (BBB) leakage, edema, inflammation, tissue structure, axon/myelin integrity, and neurotrophic factors. RESULTS: SIN-1 treatment of TBI increased whereas GSNO treatment decreased peroxynitrite, lipid peroxides/aldehydes, BBB leakage, inflammation and edema in a short-term treatment (4-48 hours). GSNO also reduced brain infarctions and enhanced the levels of NO and GSH. In a long-term treatment (14 days), GSNO protected axonal integrity, maintained myelin levels, promoted synaptic plasticity, and enhanced the expression of neurotrophic factors. CONCLUSION: Our findings indicate the participation of peroxynitrite in the pathobiology of TBI. GSNO treatment of TBI not only reduces peroxynitrite but also protects the integrity of the neurovascular unit, indicating that GSNO blunts the deleterious effects of peroxynitrite. A long-term treatment of TBI with the same low dose of GSNO promotes synaptic plasticity and enhances the expression of neurotrophic factors. These results support that GSNO reduces the levels of oxidative metabolites, protects the neurovascular unit, and promotes neurorepair mechanisms in TBI.


Brain Injuries/drug therapy , Nerve Regeneration/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Oxidative Stress/drug effects , S-Nitrosoglutathione/pharmacology , S-Nitrosoglutathione/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain/anatomy & histology , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Glutathione/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lipid Peroxidation , Male , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Donors/administration & dosage , Peroxynitrous Acid/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Receptor, trkB/genetics , Receptor, trkB/metabolism , S-Nitrosoglutathione/administration & dosage , Synaptophysin/genetics , Synaptophysin/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
15.
J Physiol Biochem ; 67(4): 539-49, 2011 Dec.
Article En | MEDLINE | ID: mdl-21626204

The alteration and further damage of vascular smooth muscle function have been implicated in the development of vascular complications and diabetes. Little is known about protein tyrosine nitration in vascular smooth muscle cell injury induced by high glucose. In this article, vascular smooth muscle cell was exposed to 30 and 40 mM high glucose for 72 h, and then the cell injury in vascular smooth muscle cell induced by high glucose was studied. It was found that high glucose stimulated vascular smooth muscle cell injury in a dose-dependent manner, including decreasing intracellular and extracellular glutathione contents, increasing malondialdehyde and intracellular reactive oxygen species content, increasing the production of nitric oxide (increased nitrite content in cell and medium), as well as increasing protein tyrosine nitration. By comparing protein tyrosine nitration induced by high glucose conditions and extrinsic factors (hemin-nitrite-glucose oxidase system and 3-morpholinosydnonimine), it may be speculated that protein is nitrated selectively, and specific protein tyrosine nitration is involved in diabetic vascular complications.


Glucose/administration & dosage , Lipid Peroxidation/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Animals , Aorta/drug effects , Aorta/metabolism , Cell Survival/drug effects , Glucose Oxidase/administration & dosage , Glutathione/metabolism , Hemin/administration & dosage , Male , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nitric Oxide/administration & dosage , Primary Cell Culture , Rats , Rats, Wistar , Tyrosine/drug effects , Tyrosine/metabolism
16.
Free Radic Biol Med ; 50(11): 1461-70, 2011 Jun 01.
Article En | MEDLINE | ID: mdl-21295134

Superoxide has been shown to play a major role in ventricular remodeling and arrhythmias after myocardial infarction. However, the source of increased myocardial superoxide production and the role of superoxide in sympathetic innervation remain to be further characterized. Male Wistar rats, after coronary artery ligation, were randomized to vehicle, allopurinol, or apocynin for 4weeks. To determine the role of peroxynitrite in sympathetic reinnervation, we also used 3-morpholinosydnonimine (a peroxynitrite generator). The postinfarction period was associated with increased oxidative stress, as measured by myocardial superoxide, nitrotyrosine, xanthine oxidase activity, NADPH oxidase activity, and dihydroethidium fluorescent staining. Measurement of myocardial norepinephrine levels revealed a significant elevation in vehicle-treated infarcted rats compared with sham. Sympathetic hyperinnervation was blunted after administration of allopurinol. Arrhythmic scores in the allopurinol-treated infarcted rats were significantly lower than those in vehicle. For similar levels of ventricular remodeling, apocynin had no beneficial effects on oxidative stress, sympathetic hyperinnervation, or arrhythmia vulnerability. Allopurinol-treated hearts had significantly decreased nerve growth factor expression, which was substantially increased after coadministration of 3-morpholinosydnonimine. These results indicate that xanthine oxidase but not NADPH oxidase largely mediates superoxide production after myocardial infarction. Xanthine oxidase inhibition ameliorates sympathetic innervation and arrhythmias possibly via inhibition of the peroxynitrite-mediated nerve growth factor pathway.


Myocardial Infarction/drug therapy , Myocardium/metabolism , NADPH Oxidases/antagonists & inhibitors , Sympathetic Nervous System/drug effects , Xanthine Oxidase/antagonists & inhibitors , Acetophenones/pharmacology , Acetophenones/therapeutic use , Allopurinol/pharmacology , Allopurinol/therapeutic use , Animals , Arrhythmias, Cardiac/prevention & control , Coronary Vessels/drug effects , Coronary Vessels/pathology , Coronary Vessels/surgery , Ethidium/analogs & derivatives , Male , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Nerve Growth Factor/genetics , Oxidative Stress/drug effects , Peroxynitrous Acid/metabolism , Rats , Rats, Wistar , Superoxides/metabolism , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/pathology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Am J Physiol Heart Circ Physiol ; 299(6): H2037-45, 2010 Dec.
Article En | MEDLINE | ID: mdl-20852057

We recently developed a rat model of cardiorenal failure that is characterized by severe left ventricular systolic dysfunction (LVSD) and low nitric oxide (NO) production that persisted after temporary low-dose NO synthase inhibition. We hypothesized that LVSD was due to continued low NO availability and might be reversed by supplementing NO. Rats underwent a subtotal nephrectomy and were treated with low-dose NO synthase inhibition with N(ω)-nitro-l-arginine up to week 8. After 3 wk of washout, rats were treated orally with either the long-acting, tolerance-free NO donor molsidomine (Mols) or vehicle (Veh). Cardiac and renal function were measured on weeks 11, 13, and 15. On week 16, LV hemodynamics and pressure-volume relationships were measured invasively, and rats were killed to quantify histological damage. On week 15, blood pressure was mildly reduced and creatinine clearance was increased by Mols (both P < 0.05). Mols treatment improved ejection fraction (53 ± 3% vs. 37 ± 2% in Veh-treated rats, P < 0.001) and stroke volume (324 ± 33 vs. 255 ± 15 µl in Veh-treated rats, P < 0.05). Rats with Mols treatment had lower end-diastolic pressures (8.5 ± 1.1 mmHg) than Veh-treated rats (16.3 ± 3.5 mmHg, P < 0.05) and reduced time constants of relaxation (21.9 ± 1.8 vs. 30.9 ± 3.3 ms, respectively, P < 0.05). The LV end-systolic pressure-volume relationship was shifted to the left in Mols compared with Veh treatment. In summary, in a model of cardiorenal failure with low NO availability, supplementing NO significantly improves cardiac systolic and diastolic function without a major effect on afterload.


Cardiotonic Agents/pharmacology , Kidney Diseases/drug therapy , Molsidomine/pharmacology , Myocardium/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , Administration, Oral , Animals , Biomarkers/blood , Cardiotonic Agents/administration & dosage , Creatinine/blood , Disease Models, Animal , Gene Expression Regulation , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Male , Molsidomine/administration & dosage , Myocardial Contraction/drug effects , Nephrectomy , Nitric Oxide Donors/administration & dosage , Nitroarginine , Rats , Rats, Inbred Lew , Stroke Volume/drug effects , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
18.
Am J Hypertens ; 23(7): 802-8, 2010 Jul.
Article En | MEDLINE | ID: mdl-20360751

BACKGROUND: In fawn-hooded hypertensive (FHH) rats, a model of hypertension, impaired preglomerular resistance, hyperfiltration, and progressive renal injury, we recently observed that supporting perinatal nitric oxide (NO) availability with the NO donor molsidomine persistently reduced blood pressure (BP) and ameliorated renal injury in male and female offspring. However, beneficial effects of perinatal molsidomine treatment were more pronounced in female than in male FHH rats. METHODS: To evaluate whether such protective effects could also be achieved with micronutrients, and whether the gender-dependent differences could be confirmed, we tested perinatal exposure to the micronutrients L-arginine, taurine, vitamin C, and vitamin E (ATCE) in FHH rats. Perinatal micronutrients increased urinary NO metabolite, sodium and potassium excretion only at 4 weeks of age, i.e., at the end of treatment. RESULTS: From 12 weeks onwards, control males had a significantly higher systolic BP (SBP) than females (P < 0.01); however after perinatal micronutrients, this difference was no longer present, indicating a pronounced antihypertensive effect of perinatal micronutrients in males (interaction P < 0.001). Development of proteinuria was attenuated by perinatal micronutrients in males and females. However, only females showed reduced glomerular filtration rate, filtration fraction, and glomerulosclerosis (GS) after perinatal micronutrients. CONCLUSIONS: In sum, perinatal micronutrients that enhance NO availability ameliorated development of hypertension and proteinuria in FHH rats. Antihypertensive effects were more pronounced in male FHH offspring, whereas renal protective effects were more pronounced in female FHH offspring. Mechanisms underlying gender-specific consequences of perinatal micronutrients require further study.


Dietary Supplements , Hypertension/prevention & control , Kidney Diseases/prevention & control , Micronutrients/administration & dosage , Proteinuria/prevention & control , Animals , Arginine/administration & dosage , Ascorbic Acid/administration & dosage , Blood Pressure , Female , Hypertension/complications , Kidney Diseases/etiology , Kidney Diseases/pathology , Male , Molsidomine/administration & dosage , Nitric Acid/metabolism , Nitric Acid/urine , Nitric Oxide Donors/administration & dosage , Potassium/metabolism , Potassium/urine , Proteinuria/etiology , Rats , Rats, Inbred Strains , Sex Factors , Sodium/metabolism , Sodium/urine , Taurine/administration & dosage , Vitamin E/administration & dosage
19.
Cell Mol Neurobiol ; 30(1): 113-21, 2010 Jan.
Article En | MEDLINE | ID: mdl-19655242

The analgesic effect induced by opiates is often potentiated during experimental inflammatory processes. We describe here that lower doses of systemic morphine are necessary to increase thermal withdrawal latencies measured in both hind paws of mice acutely inflamed with carrageenan than in healthy ones. This bilateral potentiation seems mediated through spinal opioid receptors since it is inhibited by the intrathecal (i.t.), but not intraplantar (i.pl.) administration of the opioid receptor antagonist naloxone-methiodide, and also appears when morphine is i.t. administered. Furthermore, the i.pl. administration of the nitric oxide (NO) synthase inhibitor, L-NMMA, or the K (ATP) (+) -channel blocker, glibenclamide, to carrageenan-inflamed mice inhibits the enhanced effect of systemic morphine in the paw that receives the injection of the drug, without affecting the potentiation observed in the contralateral one. The i.pl. administration of L-NMMA also partially antagonised the analgesic effect induced by i.t. morphine in inflamed mice. Finally, the increased analgesic effect evoked by the i.pl. administration of the NO donor SIN-1 either in the inflamed or in the contralateral paw of carrageenan-inflamed mice suggests that enhanced responsiveness to the peripheral analgesic effect of NO may be also underlying the bilateral potentiation of morphine-induced analgesia in acutely inflamed mice.


Analgesia , Inflammation/pathology , Morphine/pharmacology , Spinal Cord/pathology , Acute Disease , Animals , Carrageenan , Glyburide/administration & dosage , Glyburide/pharmacology , Inflammation/chemically induced , Injections, Spinal , Mice , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , Morphine/administration & dosage , Naloxone/administration & dosage , Naloxone/analogs & derivatives , Naloxone/pharmacology , Nociceptors/metabolism , Pain/pathology , Quaternary Ammonium Compounds/administration & dosage , Quaternary Ammonium Compounds/pharmacology , Temperature , omega-N-Methylarginine/administration & dosage , omega-N-Methylarginine/pharmacology
20.
Neurosci Lett ; 459(2): 79-83, 2009 Aug 07.
Article En | MEDLINE | ID: mdl-19427360

Previous studies have showed that SIN-1, a nitric oxide (NO) donor, injected into the dorsolateral column of the periaqueductal gray (dlPAG) induces flight reactions. This drug, however, can also produce peroxynitrite, which may interfere in this effect. In addition, it is also unknown if this effect is mediated by local activation of soluble guanylate cyclase (sGC). The aims of this study, therefore, were (1) to investigate if NOC-9 (6-(2-Hydroxy-1-methyl-2-nitrosohydrazino)-N-methyl-1-hexanamine), a NO donor that does not produce peroxynitrite, would produce flight reactions after intra-dlPAG administration similar to those induced by SIN-1; (2) to verify if these responses could be prevented by local injection of a selective guanylate cyclase inhibitor (ODQ). Male Wistar rats (n=5-12) with cannulae aimed at the dlPAG received injections of TRIS (pH 10.0, 0.5 microl), NOC-9 (75 and 150 nmol), saline or SIN-1 (200 nmol) and were placed in an open arena for 10 min. In a subsequent experiment animals (n=7-8) were pretreated with ODQ (1 nmol/0.5 microl) before receiving NOC-9 150 nmol. NOC-9 induced a significant dose-dependent increase in flight reactions in the first minute after injection (% of animals displaying flight: vehicle=0%, NOC 75=67%, NOC 150=75%). SIN-1 had a similar effect (100% of animals showing flight) but the effects lasted longer (10 min) than those of NOC-9. The effect of NOC-9 (150 nmol) was prevented by pretreatment with ODQ (% of animals displaying flight: vehicle+NOC 150=71%, ODQ+NOC 150=37%). The results suggest that NO donors injected into the dlPAG induce defensive responses that are not mediated by secondary peroxynitrite production. Moreover, they also indicate that these defensive responses depend on activation of local sGC. The data strengthen the proposal that NO can modulate defensive reactions in the dlPAG.


Escape Reaction/drug effects , Guanylate Cyclase/metabolism , Nitric Oxide Donors/pharmacology , Periaqueductal Gray/drug effects , Triazenes/pharmacology , Animals , Catheterization , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Guanylate Cyclase/antagonists & inhibitors , Male , Microinjections , Molsidomine/administration & dosage , Molsidomine/analogs & derivatives , Motor Activity/drug effects , Multivariate Analysis , Oxadiazoles/administration & dosage , Quinoxalines/administration & dosage , Random Allocation , Rats , Rats, Wistar , Time Factors
...