Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
Space Sci Rev ; 215(1): 9, 2019.
Article in English | MEDLINE | ID: mdl-30880847

ABSTRACT

With the advent of the Heliophysics/Geospace System Observatory (H/GSO), a complement of multi-spacecraft missions and ground-based observatories to study the space environment, data retrieval, analysis, and visualization of space physics data can be daunting. The Space Physics Environment Data Analysis System (SPEDAS), a grass-roots software development platform (www.spedas.org), is now officially supported by NASA Heliophysics as part of its data environment infrastructure. It serves more than a dozen space missions and ground observatories and can integrate the full complement of past and upcoming space physics missions with minimal resources, following clear, simple, and well-proven guidelines. Free, modular and configurable to the needs of individual missions, it works in both command-line (ideal for experienced users) and Graphical User Interface (GUI) mode (reducing the learning curve for first-time users). Both options have "crib-sheets," user-command sequences in ASCII format that can facilitate record-and-repeat actions, especially for complex operations and plotting. Crib-sheets enhance scientific interactions, as users can move rapidly and accurately from exchanges of technical information on data processing to efficient discussions regarding data interpretation and science. SPEDAS can readily query and ingest all International Solar Terrestrial Physics (ISTP)-compatible products from the Space Physics Data Facility (SPDF), enabling access to a vast collection of historic and current mission data. The planned incorporation of Heliophysics Application Programmer's Interface (HAPI) standards will facilitate data ingestion from distributed datasets that adhere to these standards. Although SPEDAS is currently Interactive Data Language (IDL)-based (and interfaces to Java-based tools such as Autoplot), efforts are under-way to expand it further to work with python (first as an interface tool and potentially even receiving an under-the-hood replacement). We review the SPEDAS development history, goals, and current implementation. We explain its "modes of use" with examples geared for users and outline its technical implementation and requirements with software developers in mind. We also describe SPEDAS personnel and software management, interfaces with other organizations, resources and support structure available to the community, and future development plans. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s11214-018-0576-4) contains supplementary material, which is available to authorized users.

2.
Opt Express ; 24(8): 8081-7, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27137247

ABSTRACT

We present the first quantum key distribution (QKD) experiment over multicore fiber. With space division multiplexing, we demonstrate that weak QKD signals can coexist with classical data signals launched at full power in a 53 km 7-core fiber, while showing negligible degradation in performance. Based on a characterization of intercore crosstalk, we perform additional simulations highlighting that classical data bandwidths beyond 1Tb/s can be supported with high speed QKD on the same fiber.

3.
Br J Clin Pharmacol ; 59(1): 85-93, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15606445

ABSTRACT

AIMS: Interaction of phosphodiesterase type 5 inhibitors for the treatment of erectile dysfunction with organic nitrates could lead to severe hypotension. NMI 861 is a combination of 7.7 mg yohimbine tartrate and 6 g l-arginine glutamate. A similar oral combination, which contains the same amount of yohimbine and L-arginine, has been shown to improve erectile function in previous studies. METHODS: In two placebo-controlled, randomized, double-blind, two-way crossover design studies we aimed to assess first the pharmacokinetics and pharmacodynamics of a single oral dose of NMI 861 administered in 16 healthy male subjects, and then the pharmacodynamics of orally administered NMI 861 in combination with intravenous nitroglycerine (GTN) in 12 healthy male subjects. Systolic (SBP) and diastolic (DBP) blood pressures, pulse rate and adverse events were measured in each study. RESULTS: NMI 861 was well tolerated by all subjects with no significant adverse reactions reported. For L-arginine, mean C(max) +/- SEM (range) was 42 +/- 2.2 (28-63) microg ml(-1) and t(max) (range) was 0.88 (0.50-1.5) h. AUC and t(1/2) were not calculated for L-arginine because of the presence of endogenous concentrations and the contribution from food sources. For yohimbine, mean C(max) was 42 +/- 11 (2.8-128) ng ml(-1); t(max) was 0.57 (0.25-1.0) h; mean AUC(0,8 h) was 65 +/- 24 (5.4-332), ng ml(-1) h and t(1/2) was 1.0 +/- 0.34 (0.40-6.0) h. There was a small but significant difference in the mean change from baseline for SBP from 0 to 6 h after NMI 861 treatment compared with placebo (0.8 +/- 1.4 vs-4.1 +/- 2.1 mmHg, respectively; 95% CI 0.0, 9.8 mmHg (P = 0.047)). There was no significant difference in SBP between treatments for the studied periods 6-12 h and 12-24 h. There was no significant difference in DBP or pulse between NMI 861 and placebo treatments for the three studied time periods. In the study designed to investigate the interaction of organic nitrate with NMI 861, subjects were infused intravenously with increasing doses of GTN (15 min each dose) at 2.5, 5, 10, 20 and 40 microg min(-1) starting 40 min after a single oral dose of either NMI 861 or placebo. There was no significant difference in the hypotensive response induced by GTN between the NMI 861 and placebo treatments. The mean maximum changes from baseline during GTN infusion for subjects administered with either NMI 861 or placebo were a decrease of 16.9 +/- 3.4 vs 13.6 +/- 2.4 mmHg (mean difference between treatments -3.3 mmHg, 95% CI -12.7, 6.0 mmHg (P = 0.460)) for SBP, a decrease of 14.7 +/- 2.0 vs 14.0 +/- 2.0 mmHg for DBP (mean difference -0.7 mmHg, 95% CI -8.2, 6.8 mmHg (P = 0.835)), and an increase of 11.8 +/- 1.9 vs 14.1 +/- 2.4 beats min(-1) for pulse, respectively (mean difference -2.3 beats min(-1), 95% CI -9.3, 4.5 beats min(-1) (P = 0.464)). CONCLUSIONS: Acute oral administration of NMI 861 was found to be well tolerated and bioavailable in healthy male subjects and no significant hypotensive interaction with intravenous GTN was detected at the doses investigated.


Subject(s)
Adrenergic alpha-Antagonists/administration & dosage , Arginine/administration & dosage , Erectile Dysfunction/drug therapy , Vasodilator Agents/administration & dosage , Yohimbine/administration & dosage , Administration, Oral , Adolescent , Adrenergic alpha-Antagonists/pharmacokinetics , Adrenergic alpha-Antagonists/pharmacology , Adult , Arginine/pharmacokinetics , Arginine/pharmacology , Cross-Over Studies , Double-Blind Method , Drug Combinations , Drug Interactions , Humans , Infusions, Intravenous , Male , Nitroglycerin , Yohimbine/pharmacokinetics , Yohimbine/pharmacology
4.
Allergy ; 59(7): 772-80, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15180766

ABSTRACT

BACKGROUND: FcgammaRIIB are low-affinity immunoglobulin (Ig)G receptors that we previously demonstrated to negatively regulate IgE-induced mast cell activation when coaggregated with FcepsilonRI. Here, we engineered and characterized a bispecific reagent capable of coaggregating FcgammaRIIB with FcepsilonRI on human mast cells and basophils. METHODS: A bispecific antibody was constructed by chemically crosslinking one Fab' fragment against human IgE and one Fab' fragment against human FcgammaRII. This molecule was used to coaggregate FcepsilonRI with FcgammaRII on human mast cells and basophils sensitized with human IgE antibodies, and the effect of coaggregation was examined on mediator release upon challenge with specific antigen. RESULTS: When used under these conditions, this bispecific antibody not only failed to trigger the release of histamine by IgE-sensitized cells, but it also prevented specific antigen from triggering histamine release. Comparable inhibitions were observed with mast cells and basophils derived in vitro from cord blood cells and with peripheral blood basophils. CONCLUSIONS: The bispecific antibody described here is the prototype of similar molecules that could be used in new therapeutic approaches of allergic diseases based on the coaggregation of activating receptors, such as FcepsilonRI, with inhibitory receptors, such as FcgammaRIIB, that are constitutively expressed by mast cells and basophils.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antigens/immunology , Basophils/immunology , Histamine Release/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Receptors, IgG/immunology , Antibodies, Bispecific/chemistry , Antigens/physiology , Basophils/physiology , Cells, Cultured , Chromatography, High Pressure Liquid/methods , Enzyme-Linked Immunosorbent Assay/methods , Fluorescent Antibody Technique/methods , Histamine Antagonists/immunology , Histamine Antagonists/pharmacology , Histamine Release/drug effects , Humans , Immunoglobulin E/drug effects , Mast Cells/physiology , Receptor Aggregation/immunology , Receptor Aggregation/physiology , Receptors, IgE/drug effects , Receptors, IgE/immunology , Receptors, IgG/drug effects
5.
Life Sci ; 73(7): 947-52, 2003 Jul 04.
Article in English | MEDLINE | ID: mdl-12798419

ABSTRACT

The effects of the selective delta-1 (delta(1)) opioid receptor agonist, DPDPE, and the selective delta(2) opioid receptor agonist, DSLET, have been studied on the ventricular fibrillation threshold (VFT) in rats with an experimental post-infarction cardiosclerosis (CS). It has been found that CS induced a significant decrease in VFT. This CS-induced decrease in VFT was significantly reversed by intravenous administration of DPDPE (0.1 mg/kg) 10 min before VFT measurement. On the contrary, intravenous injection of DSLET (0.5 mg/kg) exacerbated the CS-induced cardiac electrical instability. Pretreatment with the selective delta opioid receptor antagonist, ICI 174,864 (0.5 mg/kg), completely abolished the changes in VFT produced by both DPDPE and DSLET. Previous administration of a nonselective peripherally acting opioid receptor antagonist, naloxone methiodide (5 mg/kg) also completely reversed the antifibrillatory action of DPDPE. Naloxone methiodide and ICI 174,864 alone had no effect on VFT. Pretreatment with the nonselective K(ATP) channel blocker, glibenclamide (0.3 mg/kg), or with the mitochondrial selective K(ATP) channel blocker, 5-hydroxydecanoic acid (5-HD, 5 mg/kg), completely abolished the DPDPE-induced increase in cardiac electrical stability. Glibenclamide and 5-HD alone had no effect on VFT. These results demonstrate that the delta opioid receptor plays an important role in the regulation of electrical stability in rats with post-infarction cardiosclerosis. We propose that peripheral delta(1) opioid receptor stimulation reverses CS-induced electrical instability via mitochondrial K(ATP) channels. On the contrary, delta(2) opioid receptor stimulation may exacerbate the CS-induced decrease in VFT. Further studies are necessary to determine the delta opioid receptor subtype which mediates the antifibrillatory effect of DPDPE and pro-fibrillatory effect of DSLET.


Subject(s)
Adenosine Triphosphate/metabolism , Enkephalin, Leucine/analogs & derivatives , Myocardial Infarction/complications , Myocardium/pathology , Naloxone/analogs & derivatives , Potassium Channels/metabolism , Receptors, Opioid, delta/metabolism , Ventricular Fibrillation/metabolism , Analgesics, Opioid/pharmacology , Animals , Decanoic Acids/pharmacology , Disease Models, Animal , Drug Antagonism , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enkephalin, Leucine/pharmacology , Glyburide/pharmacology , Hydroxy Acids/pharmacology , Male , Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Naloxone/pharmacology , Quaternary Ammonium Compounds , Rats , Rats, Wistar , Receptors, Opioid, delta/agonists , Sclerosis , Ventricular Fibrillation/drug therapy , Ventricular Fibrillation/etiology
6.
Pharmacol Ther ; 91(3): 215-43, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11744068

ABSTRACT

Although yohimbine (YOH) has been available for the treatment of male erectile dysfunction (ED) for longer than Viagra, there is a perception that little is known about the clinical performance of the drug. This review attempts, by comprehensive analysis of the literature, to cover the clinical, pharmacological, and therapeutic profiles of YOH, relevant to its potential utility in the management of patients with ED. Relatively few well-designed studies have been completed. From these, however, it can be concluded that YOH as monotherapy possesses only modest efficacy in ED patients. In acute and chronic (long-term) studies, YOH has been found to be relatively free of side effects over the dose range predicted to be effective in ED. At much higher doses, the most frequently observed effects, consistent with the primary pharmacological action of the drug, are elevation of blood pressure, a slight anxiogenic action, and increased frequency of urination. These side effects are all easily reversible on termination of YOH therapy. There is increasing evidence that the erectogenic action of YOH can be augmented by concomitant administration of agents that augment the release and/or action of nitric oxide in the corpus cavernosum. YOH has yet to be studied in female sexual dysfunction. Overall, the benefit risk profile of YOH would indicate that it has potential, more probably as part of a combination strategy, e.g., with a drug that enhances the nitric oxide pathway, in the treatment of ED.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Erectile Dysfunction/drug therapy , Yohimbine/pharmacology , Adrenergic alpha-Antagonists/adverse effects , Adrenergic alpha-Antagonists/pharmacokinetics , Anxiety/chemically induced , Clinical Trials as Topic , Drug Administration Schedule , Drug Therapy, Combination , Female , Humans , Hypertension/chemically induced , Male , Nitric Oxide/chemistry , Risk Factors , Treatment Outcome , Urination Disorders/chemically induced , Yohimbine/adverse effects , Yohimbine/pharmacokinetics
7.
Clin Chem ; 47(10): 1912-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11568118

ABSTRACT

BACKGROUND: High-density microarrays are ideally suited for analyzing thousands of genes against a small number of samples. The next step in the discovery process is to take the resulting genes of interest and rapidly screen them against thousands of patient samples, tissues, or cell lines to further investigate their involvement in disease risk or the response to medication. METHODS: We used a microarray technology platform for both single-nucleotide polymorphisms (SNPs) and protein expression. Each microarray contains up to 250 elements that can be customized for each application. Slides contained either a 16- or 96-microarray format (4000-24,000 elements per slide), allowing the corresponding number of samples to be rapidly processed in parallel. RESULTS: Results for SNP genotyping and protein profiling agreed with results of restriction fragment length polymorphism (RFLP) analysis or ELISA, respectively. Genotyping analyses, using the microarray technology, on large sample sets over multiple polymorphisms in the NAT2 gene were in full agreement with traditional methodologies, such as sequencing and RFLP analysis. The multiplexed protein microarray had correlation coefficients of 0.82-0.99 (depending on analyte) compared with ELISAs. CONCLUSIONS: The integrated microarray technology platform is adaptable and versatile, while offering the high-throughput capabilities needed for drug development and discovery applications.


Subject(s)
Genome , Oligonucleotide Array Sequence Analysis/methods , Proteome , Arylamine N-Acetyltransferase/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression , Genotype , Humans , Pharmacogenetics , Polymorphism, Single Nucleotide
8.
Vopr Med Khim ; 47(3): 338-45, 2001.
Article in Russian | MEDLINE | ID: mdl-11558316

ABSTRACT

Intravenous administration of peptide delta 2-opioid receptor agonists increased cardiac sarcolemma resistance to subsequent oxidative stress in the isolated perfused rat heart in vitro. delta-Opioid receptor stimulation prevented oxidative stress-induced accumulation of lipid peroxides in myocardium and inhibition of superoxide dismutase. The cardioprotective and "antioxidant" effects of delta-agonists were completely abolished by in vivo pretreatment with the delta-receptor antagonist ICI 174,864. mu-Opioid receptor activation in vivo did not protect the isolated myocardium from cardiotoxic action of activated oxygen species.


Subject(s)
Antioxidants/metabolism , Catalase/metabolism , Myocardial Reperfusion Injury/enzymology , Oxidative Stress , Receptors, Opioid, delta/metabolism , Superoxide Dismutase/metabolism , Analgesics, Opioid/pharmacology , Animals , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Lipid Peroxidation , Myocardial Reperfusion Injury/prevention & control , Rats , Rats, Wistar , Receptors, Opioid, delta/agonists
9.
Bull Exp Biol Med ; 131(3): 251-3, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11427912

ABSTRACT

Intravenous injection of 10 mg/kg anandamide reduces the incidence and duration of epinephrine-induced arrhythmias in rats. SR141716A and SR144528, antagonists of cannabinoid receptor I and II did not abolish the antiarrhythmic effect of anandamide. These data suggest that the antiarrhythmic effect of anandamide is nonspecific or mediated via unknown cannabinoid receptors, but not associated with activation of cannabinoid receptors I and II.


Subject(s)
Arachidonic Acids/physiology , Arrhythmias, Cardiac/chemically induced , Epinephrine/adverse effects , Heart/physiology , Receptor, Cannabinoid, CB2 , Receptors, Drug/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Camphanes/pharmacology , Electrocardiography , Endocannabinoids , Heart/drug effects , Piperidines/pharmacology , Polyunsaturated Alkamides , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptors, Cannabinoid , Receptors, Drug/antagonists & inhibitors , Rimonabant
10.
Eksp Klin Farmakol ; 64(6): 27-30, 2001.
Article in Russian | MEDLINE | ID: mdl-11871232

ABSTRACT

Preliminary administration of the delta 1-opioid receptor (delta 1-OR) peptide agonist DPDPE (0.5 mg/kg, i.v.) decreased the incidence of occlusion (10 min) and reperfusion (10 min) arrhythmias in rats. The delta 2-OR agonist DSLET did not affect arrhythmias upon the coronary artery occlusion and reperfusion. Pretreatment with the selective delta-antagonists ICI 174,864 (2.5 mg/kg) or TIPP[psi] (0.5 mg/kg) completely eliminated the antiarrhythmic effect of DPDPE. Uncapable of crossing the blood brain barrier, the nonselective OR antagonist naloxone methiodide (5 mg/kg) also abolished this effect. At the same time, hexametonium (10 mg/kg) did not antagonize the antiarrhythmic effect of DPDPE. Pretreatment with the KATP channel blocker glibenclamide (0.3 mg/kg) completely eliminated the protective effect of the delta 1-OR stimulation. It was concluded that the delta 1-OR stimulation prevents the ischemic and reperfusion arrhythmias by means of the KATP channel activation.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Potassium Channels/physiology , Receptors, Opioid, delta/agonists , Reperfusion Injury/prevention & control , Animals , Arrhythmias, Cardiac/physiopathology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Naloxone/pharmacology , Oligopeptides/pharmacology , Rats , Receptors, Opioid, delta/antagonists & inhibitors , Reperfusion Injury/physiopathology
11.
Eksp Klin Farmakol ; 63(5): 27-30, 2000.
Article in Russian | MEDLINE | ID: mdl-11109521

ABSTRACT

Intraperitoneal administration of the sigma-receptor antagonist DuP 734 (1 mg/kg) 15 min before heart excision produces a decrease in the reperfusion heart contractility and prevented from the reperfusion induced cardiac cell lesion upon global ischemia of the isolated perfused rat heart. At the same time, a preliminary treatment with DuP 734 potentiated the reperfusion induced suppression of the cardiac pump function, while affecting neither the cardiac contractility dysfunction nor the cardiac cell injury during the oxidative stress. It is concluded that DuP 734 is not effective in preventing the myocardial stunning. The cardio-protector effect of DuP 734 during reperfusion is not related to inhibition of the free radical cell damage.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Myocardial Contraction/drug effects , Oxidative Stress , Piperidines/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Reperfusion Injury/drug therapy , Creatine Kinase/metabolism , Diastole , Free Radicals/metabolism , Heart/physiopathology , In Vitro Techniques , Reperfusion Injury/physiopathology , Ventricular Pressure/drug effects
12.
J Med Chem ; 43(21): 4005-16, 2000 Oct 19.
Article in English | MEDLINE | ID: mdl-11052806

ABSTRACT

Despite its widespread use, diclofenac has gastrointestinal liabilities common to nonsteroidal antiinflammatory drugs (NSAIDs) that might be reduced by concomitant administration of a gastrointestinal cytoprotectant such as nitric oxide (NO). A series of novel diclofenac esters containing a nitrosothiol (-S-NO) moiety as a NO donor functionality has been synthesized and evaluated in vivo for bioavailability, pharmacological activity, and gastric irritation. All S-NO-diclofenac derivatives acted as orally bioavailable prodrugs, producing significant levels of diclofenac in plasma within 15 min after oral administration to mice. At equimolar oral doses, S-NO-diclofenac derivatives (20a-21b) displayed rat antiinflammatory and analgesic activities comparable to those of diclofenac in the carrageenan-induced paw edema test and the mouse phenylbenzoquinone-induced writhing test, respectively. All tested S-NO-diclofenac derivatives (20a-21b) were gastric-sparing in that they elicited markedly fewer stomach lesions as compared to the stomach lesions caused by a high equimolar dose of diclofenac in the rat. Nitrosothiol esters of diclofenac comprise a novel class of NO-donating compounds having therapeutic potential as nonsteroidal antiinflammatory agents with an enhanced gastric safety profile.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Diclofenac/chemical synthesis , Nitroso Compounds/chemical synthesis , Prodrugs/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biological Availability , Diclofenac/chemistry , Diclofenac/pharmacokinetics , Diclofenac/pharmacology , Male , Mice , Nitroso Compounds/chemistry , Nitroso Compounds/pharmacokinetics , Nitroso Compounds/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Stomach/pathology , Structure-Activity Relationship
13.
J Immunol ; 165(6): 3154-61, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975829

ABSTRACT

The plasminogen activator inhibitor type 1 (PAI-1) has an essential role in tissue remodeling. The PAI-1 gene was induced by a combination of phorbol ester and calcium ionophore at the highest level among the inducible human mast cell genes that we have analyzed on a DNA microarray. PAI-1 was secreted by both a human mast cell line (HMC)-1 and primary cultured human mast cells upon stimulation, whereas PAI-1 was undetectable in either group of unstimulated cells. The secretion of PAI-1 was due to de novo synthesis of PAI-1 rather than secretion of preformed PAI-1. The functional significance of PAI-1 secretion was demonstrated by complete inhibition of tissue-type plasminogen activator activity with supernatants of stimulated HMC-1 cells. Furthermore, we were able to regulate PAI-1 gene expression in HMC-1 cells by known therapeutic agents. High-dose (1 microM) dexamethasone induced PAI-1 mRNA expression. Cyclosporin down-regulated the expression of the PAI-1 gene. Cycloheximide abrogated PAI-1 mRNA expression, suggesting that transcription of the PAI-1 gene requires de novo synthesis of early gene products, including transcription factors. Finally, we demonstrated PAI-1 in lung mast cells from a patient with asthmatic attack by double-immunofluorescence study. This is the first report demonstrating that activated human mast cells release a striking amount of functionally active PAI-1. These results suggest that PAI-1 could play an important role in airway remodeling of asthma, and inhibition of PAI-1 activity could represent a novel therapeutic approach in the management of airway remodeling.


Subject(s)
Asthma/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Asthma/immunology , Asthma/pathology , Cells, Cultured , Cycloheximide/pharmacology , Cyclosporine/pharmacology , Dexamethasone/pharmacology , Fibrinolysis/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Kinetics , Lung/immunology , Lung/metabolism , Mast Cells/pathology , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/physiology , Plasminogen Activators/biosynthesis , Plasminogen Activators/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Receptors, IgE/immunology , Receptors, IgE/metabolism , Tissue Plasminogen Activator/antagonists & inhibitors
14.
Eksp Klin Farmakol ; 63(2): 39-43, 2000.
Article in Russian | MEDLINE | ID: mdl-10834092

ABSTRACT

It has been found that the sigma 1- and sigma 3-receptor antagonists, DuP 734 intraperitoneally and XJ 448 intravenously, had antiarrhythmic effects against epinephrine-induced arrhythmias in rats. The ED50 for antiarrhythmic effect of DuP 734 was 0.157 mg/kg after intraperitoneal administration. Other sigma-receptor antagonists (rimcazole, BMY 14802, haloperidol) did not affect the incidence of epinephrine-induced arrhythmias. sigma 1-Receptor agonists (DTG, N-allyl-normetazocine, (+)-3-PPP, (-)-3-PPP) had proarrhythmic effect after systemic administration. The sigma-agonist N-allyl-normetazocine and sigma-antagonist XJ 488 had no effect on the incidence of epinephrine-induced arrhythmias after intracerebroventricular administration. Therefore, it appears that the central nervous system does not play a significant role in the antiarrhythmic or proarrhythmic effects of sigma ligands. We hypothesize that these effects of sigma ligands might be dependent on their action on cardiac sigma receptors.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/chemically induced , Disease Models, Animal , Drug Evaluation, Preclinical , Electrocardiography/drug effects , Epinephrine , Ligands , Male , Rats , Rats, Wistar , Receptors, sigma/drug effects
15.
Mol Cell Biol Res Commun ; 4(1): 32-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11152625

ABSTRACT

Nitric oxide (NO), delivered by a single addition of S-nitrosoglutathione (GSNO, IC(50) = 60-75 microM), causes the prolonged, multi-day suppression of proliferation of asynchronous, logarithmically growing human (hCASMC, two cell strains), and porcine (porCASMC) coronary artery smooth muscle cells. The inhibition is not cytotoxic, but cytostatic and reversible. Transient exposure (>4-12 h) to GSNO is sufficient to elicit prolonged suppression, but a less than 4 h exposure produces little or no inhibition. Unlike porCASMC and rat and rabbit aortic SMC, hCASMC synthesize little cGMP in response to GSNO stimulation, suggesting loss of NO responsive guanylate cyclase in vitro. The guanylate cyclase inhibitor, ODQ, blocks the slight cGMP synthesis induced by GSNO in hCASMC, but does not prevent GSNO suppression of proliferation. These data support a cGMP independent mechanism for NO induced suppression of hCASMC proliferation which may be significant in the treatment of proliferative coronary artery diseases.


Subject(s)
Cell Division/drug effects , Coronary Vessels/drug effects , Cyclic GMP/metabolism , Glutathione/analogs & derivatives , Glutathione/pharmacology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Animals , Cells, Cultured , Coronary Vessels/cytology , Humans , Muscle, Smooth, Vascular/cytology , Rabbits , Rats , S-Nitrosoglutathione , Swine
16.
Life Sci ; 65(1): PL13-7, 1999.
Article in English | MEDLINE | ID: mdl-10403501

ABSTRACT

The effects of the extremely selective mu-opioid receptor agonist, [D-Arg2,Lys4]-dermorphin-(1-4)-amide (DALDA), the mu-opioid receptor agonist morphine, the mu/delta agonist D-Ala2, Leu5, Arg6-enkephalin (dalargin), the kappa-opioid receptor agonist spiradoline, and the sigma1-receptor antagonist DuP 734 on ventricular fibrillation threshold (VFT) was investigated in an experimental post-infarction cardiosclerosis model and an immobilization stress-induced model in rats. Both models produced a significant decrease in VFT. The postinfarction cardiosclerosis-induced decrease in VFT was significantly reversed by intravenous administration of dalargin (0.1 mg/kg), DALDA (0.1 mg/kg), or morphine HCl (1.5 mg/kg). Pretreatment with naloxone (0.2 mg/kg) completely eliminated the increase in cardiac electrical stability produced by DALDA. Both spiradoline (8 mg/kg, i.p.) and DuP 734 (1 mg/kg, i.p.) produced a significant increase in VFT in rats with post-infarction cardiosclerosis. This effect of spiradoline was blocked by nor-binaltorphimine. The immobilization stress-induced decrease in VFT was significantly reversed by administration of either DALDA, spiradoline or DuP 734. In conclusion, activation of either mu- or kappa1-opioid receptors or blockade of sigma1-receptors reversed the decrease in VFT in both cardiac compromised models. Since DALDA and dalargin essentially do not cross blood brain barriers, their effects on VFT may be mediated through peripheral mu-opioid receptors.


Subject(s)
Heart/physiology , Myocardial Infarction/physiopathology , Myocardium/pathology , Receptors, Opioid, delta/physiology , Receptors, Opioid/physiology , Stress, Physiological/physiopathology , Animals , Anti-Arrhythmia Agents/pharmacology , Disease Models, Animal , Dynorphins/pharmacology , Enkephalin, Leucine-2-Alanine/analogs & derivatives , Enkephalin, Leucine-2-Alanine/pharmacology , Heart/drug effects , Heart/physiopathology , Immobilization , Ligands , Morphine/pharmacology , Myocardial Infarction/drug therapy , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists , Oligopeptides/antagonists & inhibitors , Oligopeptides/pharmacology , Piperidines/pharmacology , Pyrrolidines/antagonists & inhibitors , Pyrrolidines/pharmacology , Rats , Receptors, Opioid/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Ventricular Fibrillation/drug therapy , beta-Endorphin/pharmacology
17.
J Pharmacol Exp Ther ; 290(1): 121-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381767

ABSTRACT

We designed and evaluated a new class of molecules, nitrosylated alpha-adrenergic receptor antagonists, as potential agents for the treatment of impotence. In in vitro studies with human and rabbit corpus cavernosum strips in organ chambers, the alpha-adrenergic receptor antagonists (alpha-ARAs) moxisylyte and yohimbine and their corresponding nitrosylated compounds, SNO-moxisylyte (NMI-221) and SNO-yohimbine (NMI-187), concentration-dependently relaxed endothelin-induced contraction. The nitrosylated compounds were significantly more potent than the parent alpha-ARA. In human tissues, the specific phosphodiesterase type 5 inhibitor zaprinast potentiated the relaxing effects of the nitrosylated compounds. Only nitrosylated compounds induced accumulation of cyclic GMP in rabbit corpus cavernosum strips. Yohimbine and NMI-187 demonstrated a potent alpha2-blocking activity, with no significant differences in pA2 values (8.9 versus 8.2, respectively). Moxisylyte and NMI-221 showed moderate potency in antagonizing phenylephrine contraction, with comparable pA2 values for both molecules (6.5 versus 6.6, respectively). alpha-Adrenergic receptor-binding studies showed similar binding affinities for the alpha-ARA and their corresponding nitrosylated compounds. In vivo, intracavernosal injection of nitrosylated molecules caused greater increases in intracavernosal pressure (NMI-221 versus moxisylyte) that were more long lasting than those of moxisylyte or yohimbine. There were no significant differences between nitrosylated and non-nitrosylated compounds in the magnitude of systemic mean arterial pressure decrease after intracavernosal injection. alpha-ARA and the nitrosylated compounds showed no pain-inducing activity as evaluated with the paw-lick model in mice. In summary, nitrosylated alpha-ARA have the dual functionalities of nitric oxide donors and alpha-ARA. These drugs induced penile erection in animals, suggesting their possible therapeutic value as agents for the local pharmacological treatment of impotence.


Subject(s)
Adrenergic alpha-Antagonists/chemical synthesis , Erectile Dysfunction/drug therapy , Moxisylyte/analogs & derivatives , Nitroso Compounds/chemical synthesis , Nitroso Compounds/pharmacology , Vasodilator Agents/chemical synthesis , Yohimbine/analogs & derivatives , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Cyclic GMP/metabolism , Drug Design , Endothelins/pharmacology , Humans , In Vitro Techniques , Male , Membranes , Mice , Moxisylyte/chemical synthesis , Moxisylyte/metabolism , Moxisylyte/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Penis/drug effects , Penis/metabolism , Penis/physiology , Phenylephrine/pharmacology , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology , Yohimbine/chemical synthesis , Yohimbine/metabolism , Yohimbine/pharmacology
18.
J Med Chem ; 42(5): 805-18, 1999 Mar 11.
Article in English | MEDLINE | ID: mdl-10072679

ABSTRACT

Screening of our chemical library using a rat corticotropin-releasing hormone (CRH) receptor assay led to the discovery that 2-anilinopyrimidine 15-1 weakly displaced [125I]-0-Tyr-oCRH from rat frontal cortex homogenates when compared to the known peptide antagonist alpha-helical CRH(9-41) (Ki = 5700 nM vs 1 nM). Furthermore, 15-1 weakly inhibited CRH-stimulated adenylate cyclase activity in the same tissue, but it was less potent than alpha-helical CRH(9-41) (IC50 = 20 000 nM vs 250 nM). Systematic structure-activity relationship studies, using the cloned human CRH1 receptor assay, defined the pharmacophore for optimal binding to hCRH1 receptors. Several high-affinity 2-anilinopyrimidines and -triazines were discovered, some of which had superior pharmacokinetic profiles in the rat. This paper describes the structure-activity studies which improved hCRH1 receptor binding affinity and pharmacokinetic parameters in the rat. Compound 28-17 (mean hCRH1 Ki = 32 nM) had a significantly improved pharmacokinetic profile in the rat (19% oral bioavailability at 30 mg/kg) as well as in the dog (20% oral bioavailability at 5 mg/kg) relative to the early lead structures.


Subject(s)
Pyrimidines/chemical synthesis , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Triazines/chemical synthesis , Animals , Biological Availability , Dogs , Frontal Lobe/metabolism , Humans , In Vitro Techniques , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Receptors, Corticotropin-Releasing Hormone/metabolism , Recombinant Proteins/metabolism , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacokinetics , Triazines/pharmacology
19.
Ross Fiziol Zh Im I M Sechenova ; 85(11): 1396-408, 1999 Nov.
Article in Russian | MEDLINE | ID: mdl-10687172

ABSTRACT

Pre-treatment of the sigma-receptor with the sigma-receptor agonist (+)-SKF 10.047 improved the reperfusion recovery of cardiac pump function. The sigma-receptor activation, among other effects, prevented the reperfusion contracture, increased pressure in the left ventricle, and improved survival of cardiomyocytes after ischemia/reperfusion. Pre-treatment with the sigma-receptor antagonist DuP 734 augmented the reperfusion systolic dysfunction of the myocardium and prevented postischemic contractures and cardiac cell lesions. Activation of the cardiac sigma-receptor seems to prompt an augmentation of tolerance to the reperfusion damage.


Subject(s)
Heart/physiopathology , Myocardial Reperfusion Injury/physiopathology , Receptors, sigma/physiology , Animals , Blood Pressure , Creatine Kinase/metabolism , Electrocardiography , Heart/drug effects , Heart Rate/drug effects , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Phenazocine/analogs & derivatives , Phenazocine/pharmacology , Piperidines/pharmacology , Rats , Rats, Wistar , Receptors, sigma/agonists , Receptors, sigma/antagonists & inhibitors , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
20.
Bioorg Med Chem ; 7(11): 2321-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10632042

ABSTRACT

Structure-activity studies around the 4-position of 2-anilinopyrimidine corticotropin releasing factor (CRF) antagonists suggest that there is a large lipophilic cavity in the rat CRF receptor, which can accommodate a wide variety of substituents at this position in contrast to the steric constraints observed for other positions on the 2-anilinopyrimidine core. The chemical syntheses and biological activities of 2-anilinopyrimidine CRF antagonists with carbon-linked substituents at the 4-position are reported. Significant improvements in rat pharmacokinetic parameters were achieved relative to those for the lead structure. While the lead compound 1 (rCRF Ki = 44 nM) afforded no detectable rat plasma levels after intraperitoneal (i.p.) or oral (p.o.) dosing, compounds 3-3 (rCRF Ki = 16 nM) and 3-4 (rCRF Ki 59 nM) gave high rat plasma levels at 30 mg/kg (i.p., p.o.) (Cmax = 1389 nM and 8581 nM (i.p.) respectively; Cmax = 113 nM and 988 nM (p.o.), respectively). Furthermore 3-3 and 3-4 had superior bioavailabilities at these doses (59 and 46% (i.p.), respectively; 2 and 10%, (p.o.), respectively).


Subject(s)
Aniline Compounds/pharmacology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Pyrimidines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Adenylyl Cyclase Inhibitors , Animals , Corticotropin-Releasing Hormone/metabolism , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL