Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Phys Chem Lett ; 11(22): 9659-9668, 2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33135884

ABSTRACT

SARS-CoV-2 is the cause of the ongoing Coronavirus disease 19 (COVID-19) pandemic around the world causing pneumonia and lower respiratory tract infections. In understanding the SARS-CoV-2 pathogenicity and mechanism of action, it is essential to depict the full repertoire of expressed viral proteins. The recent biological studies have highlighted the leader protein Nsp1 of SARS-CoV-2 importance in shutting down the host protein production. Besides, it still enigmatic how Nsp1 regulates for translation. Here we report the novel structure of Nsp1 from SARS-CoV-2 in complex with the SL1 region of 5'UTR of SARS-CoV-2, and its factual interaction is corroborated with enzyme kinetics and experimental binding affinity studies. The studies also address how leader protein Nsp1 of SARS-CoV-2 recognizes its self RNA toward translational regulation by further recruitment of the 40S ribosome. With the aid of molecular dynamics and simulations, we also demonstrated the real-time stability and functional dynamics of the Nsp1/SL1 complex. The studies also report the potential inhibitors and their mode of action to block viral protein/RNA complex formation. This enhance our understanding of the mechanism of the first viral protein Nsp1 synthesized in the human cell to regulate the translation of self and host. Understanding the structure and mechanism of SARS-CoV-2 Nsp1 and its interplay with the viral RNA and ribosome will open the arena for exploring the development of live attenuated vaccines and effective therapeutic targets for this disease.


Subject(s)
5' Untranslated Regions , RNA, Viral/metabolism , SARS-CoV-2/chemistry , Viral Nonstructural Proteins/metabolism , COVID-19 Vaccines , Depsides/chemistry , Depsides/metabolism , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/metabolism , Lactones/chemistry , Lactones/metabolism , Molecular Dynamics Simulation , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Protein Binding/drug effects , RNA, Viral/chemistry , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/metabolism , SARS-CoV-2/pathogenicity , Salicylates/chemistry , Salicylates/metabolism , Viral Nonstructural Proteins/chemistry , Virulence
2.
Steroids ; 137: 1-13, 2018 09.
Article in English | MEDLINE | ID: mdl-30031853

ABSTRACT

New 3,20-dihydroxy-13α-19-norpregna-1,3,5(10)-trienes were synthesized. The effects of these compounds on breast cancer cells and ERα activation were investigated. The scaffold of compounds containing the six-membered ring D' annulated at 16α,17α-positions was constructed via the Lewis acid catalyzed Diels-Alder reaction of butadiene with 3-methoxy-13α-19-norpregna-1,3,5(10),16-tetraen-20-one 5 under a pressure of 600 MPa. The hydrogenation of primary cyclohexene adduct 6 followed by the one-pot reduction-demethylation (DIBAH) gave target epimeric 3,20-dihydroxy steroids 8a and 8b. The Corey-Chaykovsky reaction of the same conjugated ketone 5 gave a 16α,17α-methylene-substituted compound. The reaction of the latter with DIBAH yielded 3,20(R,S)-dihydroxy-16α,17α-methyleno-13α-19-norpregna-1,3,5(10)-triene 10. The hydrogenation of the 16,17-double bond of compound 5 produced a mixture of 17α- and 17ß-epimeric ketones, reduction-demethylation of which gave 3,20(S)-dihydroxy-13α,17α-19-norpregna-1,3,5(10)-triene 12a and 3,20(R)-dihydroxy-13α,17ß-19-norpregna-1,3,5(10)-triene 12b. All compounds were fully characterized by 1D and 2D NMR, HRMS, and X-ray diffraction. All target compounds showed pronounced cytotoxic effect against MCF-7 breast cancer cells and NCI/ADR-RES doxorubicin-resistant cells at micromolar concentrations. The ERα-mediated luciferase reporter gene assay demonstrated that all compounds, except for compound 10, are ERα inhibitors, while cyclopropane compound 10 proved to be an ERα activator. Docking experiments showed that all compounds are well accommodated to LBD ERα but have some differences in the binding mode.


Subject(s)
Pregnatrienes/chemical synthesis , Pregnatrienes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Chemistry Techniques, Synthetic , Estrogen Antagonists/chemical synthesis , Estrogen Antagonists/chemistry , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Estrogens/chemical synthesis , Estrogens/chemistry , Estrogens/metabolism , Estrogens/pharmacology , Humans , MCF-7 Cells , Molecular Docking Simulation , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Protein Domains , Stereoisomerism
3.
PLoS One ; 4(2): e4309, 2009.
Article in English | MEDLINE | ID: mdl-19190754

ABSTRACT

Since P450scc transforms 7-dehydrocholesterol (7DHC) to 7-dehydropregnenolone (7DHP) in vitro, we investigated sequential 7DHC metabolism by adrenal glands ex vivo. There was a rapid, time- and dose-dependent metabolism of 7DHC by adrenals from rats, pigs, rabbits and dogs with production of more polar 5,7-dienes as detected by RP-HPLC. Based on retention time (RT), UV spectra and mass spectrometry, we identified the major products common to all tested species as 7DHP, 22-hydroxy-7DHC and 20,22-dihydroxy-7DHC. The involvement of P450scc in adrenal metabolic transformation was confirmed by the inhibition of this process by DL-aminoglutethimide. The metabolism of 7DHC with subsequent production of 7DHP was stimulated by forscolin indicating involvement of cAMP dependent pathways. Additional minor products of 7DHC metabolism that were more polar than 7DHP were identified as 17-hydroxy-7DHP (in pig adrenals but not those of rats) and as pregna-4,7-diene-3,20-dione (7-dehydroprogesterone). Both products represented the major identifiable products of 7DHP metabolism in adrenal glands. Studies with purified enzymes show that StAR protein likely transports 7DHC to the inner mitochondrial membrane, that 7DHC can compete effectively with cholesterol for the substrate binding site on P450scc and that the catalytic efficiency of 3betaHSD for 7DHP (V(m)/K(m)) is 40% of that for pregnenolone. Skin mitochondria are capable of transforming 7DHC to 7DHP and the 7DHP is metabolized further by skin extracts. Finally, 7DHP, its photoderivative 20-oxopregnacalciferol, and pregnenolone exhibited biological activity in skin cells including inhibition of proliferation of epidermal keratinocytes and melanocytes, and melanoma cells. These findings define a novel steroidogenic pathway: 7DHC-->22(OH)7DHC-->20,22(OH)(2)7DHC-->7DHP, with potential further metabolism of 7DHP mediated by 3betaHSD or CYP17, depending on mammalian species. The 5-7 dienal intermediates of the pathway can be a source of biologically active vitamin D3 derivatives after delivery to or production in the skin, an organ intermittently exposed to solar radiation.


Subject(s)
Adrenal Glands/metabolism , Alkenes/metabolism , Dehydrocholesterols/metabolism , Skin/metabolism , Alkenes/chemistry , Animals , Cell Proliferation , Chromatography, Liquid , Dehydrocholesterols/chemistry , Enzyme Inhibitors , Enzymes/metabolism , Female , Humans , Keratinocytes/metabolism , Male , Mass Spectrometry , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Metabolic Networks and Pathways , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Secosteroids/chemistry , Secosteroids/metabolism , Skin/cytology , Time Factors , Tissue Extracts/metabolism
4.
Biochemistry ; 47(29): 7648-62, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18578507

ABSTRACT

The transcriptional activity of steroid hormones is intimately associated with their structure. Deacylcortivazol (DAC) contains several features that were predicted to make it an inactive glucocorticoid. Nevertheless, gene induction and repression by complexes of glucocorticoid receptor (GR) with DAC occur with potency (lower EC 50) greater than and efficacy (maximal activity, or A max) equal to those of the very active and smaller synthetic glucocorticoid dexamethasone (Dex). Guided by a recent X-ray structure of DAC bound to the GR ligand binding domain (LBD), we now report that several point mutants in the LBD have little effect on the binding of either agonist steroid. However, these same mutations dramatically alter the A max and/or EC 50 of exogenous and endogenous genes in a manner that depends on steroid structure. In some cases, Dex is no longer a full agonist. These properties appear to result from a preferential inactivation of the AF2 activation domain in the GR LBD of Dex-bound, but not DAC-bound, receptors. The Dex-bound receptors display normal binding to, but a greatly reduced response to, the coactivator TIF2, thus indicating a defect in the transmission efficiency of GR-steroid complex information to the coactivator TIF2. In addition, all GR mutants that are active in gene induction with either Dex or DAC have greatly reduced activity in gene repression. This contrasts with the reports of GR mutations preferentially suppressing GR-mediated induction. The properties of these GR mutants in gene induction support the hypothesis that the A max and EC 50 of GR-controlled gene expression can be independently modified, indicate that the receptor can be modified to favor activity with a specific agonist steroid, and suggest that new ligands with suitable substituents may be able to affect the same LBD conformational changes and thereby broaden the therapeutic applications of glucocorticoid steroids.


Subject(s)
Point Mutation , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Steroids/metabolism , Animals , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Dexamethasone/chemistry , Dexamethasone/metabolism , Gene Expression/drug effects , Humans , Mutagenesis, Site-Directed , Nuclear Receptor Coactivator 2/pharmacology , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary , Receptors, Glucocorticoid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship
5.
Steroids ; 72(9-10): 673-81, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606285

ABSTRACT

One goal of steroid research is precise differential regulation of gene expression by steroid hormone receptors through use of distinct ligands which modulate defined sets of cellular effects. Such "selective modulator" ligands are known for several receptors. Potent pyrazolo-glucocorticoid (11beta,16alpha)-21-(Acetyloxy)-11,17-dihydroxy-6,16-dimethyl-2'-phenyl-2'H-pregna-2,4,6-trieno[3,2-c]pyrazol-20-one) cortivazol activates the glucocorticoid receptor to regulate gene expression and can bring about apoptosis of leukemic CEM cells resistant to (9-fluoro-11,17-dihydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,11,12,14,15,16-octahydrocyclopenta[a]phenanthren-3-one) dexamethasone. We therefore tested the hypothesis that cortivazol and dexamethasone regulate non-identical sets of genes in CEM cells. We found that while cortivazol and dexamethasone overlap in regulation of most genes, each steroid regulates an exclusive set of transcripts in clone CEM-C7-14 (sensitive to apoptosis by both dexamethasone and cortivazol) and clone CEM-C1-15 (dexamethasone-resistant but cortivazol-sensitive). Fifty-seven genes were regulated uniquely to a statistically significant extent by cortivazol in both clones. Many of the cortivazol specific genes are key components of various signal transduction pathways. Our data clearly show cortivazol to be a selective modulator of GR action.


Subject(s)
Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Mifepristone/pharmacology , Pregnatrienes/pharmacology , Receptors, Glucocorticoid/agonists , Apoptosis/drug effects , Cell Line , Dexamethasone/chemistry , Dexamethasone/metabolism , Glucocorticoids/metabolism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Receptors, Glucocorticoid/metabolism
6.
Mol Endocrinol ; 19(5): 1110-24, 2005 May.
Article in English | MEDLINE | ID: mdl-15677712

ABSTRACT

Recent structural analyses of the nuclear receptors establish a paradigm of receptor activation, in which agonist binding induces the ligand binding domain (LBD)/activation function-2 helix to form a charge clamp for coactivator recruitment. However, these analyses have not sufficiently addressed the mechanisms for differential actions of various synthetic steroids in terms of fine tuning of multiple functions of whole receptor molecules. In the present study, we used the glucocorticoid receptor (GR)-specific agonist cortivazol (CVZ) to probe the plasticity and functional modularity of the GR. Structural docking analysis revealed that although CVZ is more bulky than other agonists, it can be accommodated in the ligand binding pocket of the GR by reorientation of several amino acid side chains but without major alterations in the active conformation of the LBD. In this induced fit model, the phenylpyrazole A-ring of CVZ establishes additional contacts with helices 3 and 5 of the LBD that may contribute to a more stable LBD configuration. Structural and functional analysis revealed that CVZ is able to compensate for the deleterious effects of a C-terminal deletion of the LBD in a manner that mimics the stabilizing influence of the F602S point mutation. CVZ-mediated productive recruitment of transcriptional intermediary factor 2 to the C-terminally deleted LBD requires the receptor's own DNA binding domain and is positively influenced by the N-terminal regions of GR or progesterone receptor. These results support a model where ligand-dependent conformational changes in the LBD play a role in GR-mediated gene regulation via modular interaction with the DBD and activation function-1.


Subject(s)
Pregnatrienes/pharmacology , Receptors, Glucocorticoid/agonists , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Dexamethasone/metabolism , Dexamethasone/pharmacology , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , HeLa Cells , Hot Temperature , Humans , Ligands , Molecular Sequence Data , NF-kappa B/metabolism , Pregnatrienes/metabolism , Protein Structure, Tertiary , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Time Factors
7.
J Steroid Biochem Mol Biol ; 87(4-5): 319-25, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14698213

ABSTRACT

The conversion of 21-acetoxy-pregna-4(5),9(11),16(17)-triene-21-ol-3,20-dione (I) by Nocardioides simplex VKM Ac-2033D was studied purposed selective production of its 1(2)-dehydroanalogues-value precursors in the synthesis of modern glucocorticoids starting from 9alpha-hydroxyandrostenes. 21-Acetoxy-pregna-1(2),4(5),9(11),16(17)-tetraene-21-ol-3,20-dione (II), pregna-4(5),9(11),16(17)-triene-21-ol-3,20-dione (III) and pregna-1(2),4(5),9(11),16(17)-tetraene-21-ol-3,20-dione (IV) were revealed as metabolites, and the structures were confirmed by mass spectrometry and (1)H nuclear magnetic resonance (NMR) spectroscopy. The metabolic pathways of I by N. simplex included 1(2)-dehydrogenation and deacetylation. The sequence of the reactions was shown to depend on the transformation conditions. The presence of both soluble and membrane associated steroid esterases in N. simplex was demonstrated using cell fractionation. Unlike inducible 1(2)-dehydrogenase, steroid esterase was shown to be constitutive. The conditions providing selective accumulation of II from I by whole N. simplex cells were determined.


Subject(s)
Nocardiaceae/metabolism , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Acetylation , Biomass , Biotransformation , Chromatography, Thin Layer , Cyclodextrins/chemistry , Cyclodextrins/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry/methods , Nocardiaceae/cytology , Nocardiaceae/genetics , Phenazines/pharmacology , Subcellular Fractions/metabolism , Time Factors
8.
J Biol Chem ; 277(7): 5529-40, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11741935

ABSTRACT

Ligand-receptor coupling is one of the important constituents of signal transduction and is essential for physiological transmission of actions of endogenous substances including steroid hormones. However, molecular mechanisms of the redundancy between glucocorticoid and mineralocorticoid actions remain unknown because of complicated cross-talk among, for example, these adrenal steroids, their cognate receptors, and target genes. Receptor-specific ligand that can distinctly modulate target gene expression should be developed to overcome this issue. In this report, we showed that a pyrazolosteroid cortivazol (CVZ) does not induce either nuclear translocation or transactivation function of the mineralocorticoid receptor (MR) but does both for the glucocorticoid receptor (GR). Moreover, deletion analysis of the C-terminal end of the GR has revealed that CVZ interacts with the distinct portion of the ligand binding domain (LBD) and differentially modulates the ligand-dependent interaction between transcription intermediary factor 2 and the LBD when compared with cortisol, dexamethasone, and aldosterone. Thus, it is indicated that CVZ may not be only a molecular probe for the analysis of the redundancy between the GR and MR in vivo but also a useful reagent to clarify structure-function relationship of the GR LBD.


Subject(s)
Glucocorticoids/chemistry , Glucocorticoids/metabolism , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Receptors, Glucocorticoid/chemistry , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Blotting, Western , CHO Cells , COS Cells , Cell Line , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Deletion , Genes, Reporter , Green Fluorescent Proteins , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Kinetics , Ligands , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Glucocorticoid/metabolism , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Structure-Activity Relationship , Time Factors , Transcriptional Activation , Transfection
9.
J Spinal Cord Med ; 24(3): 142-6, 2001.
Article in English | MEDLINE | ID: mdl-11585231

ABSTRACT

Most acute spinal cord injuries (SCI) do not involve complete transection of the spinal cord; typically, a rim of white matter survives. The potential for neurological recovery depends on optimal preservation of the ascending and descending white matter axons and their normal myelination. Pharmacologic strategies focus on the control of secondary injury processes, primarily lipid peroxidation (LP), and the salvage of as much white matter as possible. The first effective neuroprotective agent was methylprednisolone (MP), a glucocorticosteroid that in high doses improves neurological recovery in animals and humans following acute SCI. Tirilazad is a more targeted non-glucocorticoid LP inhibitor that has been shown to be neuroprotective and has fewer side effects than MP. Future SCI therapy is likely to encompass various neuroprotective agents, including inhibitors of LP, inhibitors of the nitric oxide-derived reactive oxygen species peroxynitrite, inhibitors of calpain (which is responsible for degrading the spinal cord cytoskeleton), and inhibitors of post-traumatic apoptosis of neurons and myelin-forming oligodendroglia. In addition, neuroprotective strategies will eventually be followed by neurorestorative agents that stimulate the plasticity of surviving neural pathways, and will be used in conjunction with other neurorestorative therapies like cell transplantation and gene therapy techniques.


Subject(s)
Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , Acute Disease , Humans , Lipid Peroxidation/physiology , Methylprednisolone/metabolism , Methylprednisolone/therapeutic use , Neuroprotective Agents/metabolism , Pregnatrienes/metabolism , Pregnatrienes/therapeutic use , Spinal Cord Injuries/metabolism
10.
Xenobiotica ; 27(11): 1131-45, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9413917

ABSTRACT

1. We have determined that 2,4-dipyrrolidinylpyrimidine (2,4-DPP), used as a model for studies of the metabolism of therapeutic agents containing this moiety, undergoes three characteristic hydroxylations when incubated with male rat liver microsomes. Analysis of microsomal incubates of stable isotope labelled analogues of 2,4-DPP by particle beam-liquid chromatography-mass spectrometry (LC-PB-MS) has shown that the three metabolites are 4-(3-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M1), 4-(2-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M2) and 2-(2-hydroxypyrrolidinyl)-4-(pyrrolidinyl)-pyrimidine (M3). 2. We determined that enzymes of the cytochrome P450 family are responsible for the in vitro hydroxylations of 2,4-DPP. 3. We observed that in microsomal incubations carried out in the presence of cyanide, a single cyanide adduct is formed implicating an iminium ion intermediate in the oxidation of the 2-pyrrolidine ring. 4. We also determined the intermolecular deuterium isotope effects for the formation of each of the three products. For M1, kH/kD = 14.55 +/- 0.54; for M2, kH/kD = 6.01 +/- 0.65; and for M3, kH/kD = 5.35 +/- 1.18. 5. We interpret these data as suggesting that M2 and M3 are formed by the same mechanism, probably including the formation of an iminium ion, and that M1 is formed by direct hydrogen abstraction.


Subject(s)
Antioxidants/metabolism , Cytochrome P-450 Enzyme System/metabolism , Free Radical Scavengers , Pregnatrienes/metabolism , Pyrimidines/metabolism , Pyrroles/metabolism , Animals , Biotransformation , Chromatography, Liquid , Cyanides/metabolism , Deuterium , Hydroxylation , Male , Mass Spectrometry , Oxidation-Reduction , Pregnatrienes/pharmacokinetics , Rats , Rats, Sprague-Dawley
11.
Crit Care Med ; 25(4): 652-6, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9142031

ABSTRACT

OBJECTIVE: To determine whether pretreatment with a 21-aminosteroid, U-74389G, can prevent subsequent DNA injury in bleomycin-exposed lungs. SUBJECTS: Thirty-six adult male Sprague-Dawley rats. DESIGN: Controlled animal laboratory investigation of DNA injury in vivo. INTERVENTIONS: Animals were treated with 21-aminosteroid (10 mg/kg) or vehicle and subsequently received intratracheal instillation of bleomycin (1.75 U) or normal saline. MEASUREMENTS AND MAIN RESULTS: Twenty-four hours after bleomycin exposure, the 21-aminosteroid-treated animals had decreased evidence of DNA injury, expressed as percentage of DNA fragmentation normalized to the control group (113.5 +/- 6 [SEM] vs. 132 +/- 3.9%, p < or = .05), and activity of the DNA repair enzyme poly ADP-ribose synthetase (3.4 +/- 0.2 vs. 5.6 +/- 0.9 pmol nicotinamide adenine dinucleotide/min/mg protein, p < or = .05). Only bleomycin-exposed (+ vehicle) animals demonstrated significant evidence of increased DNA injury vs. the intratracheal saline-exposed control groups. CONCLUSIONS: The 21-aminosteroid pretreatment decreases subsequent pulmonary DNA injury induced by bleomycin exposure. This finding is likely due to the 21-aminosteroid's iron-chelating and cell-permeating abilities, and suggests that these agents may be effective in other diseases where iron-dependent free radical reactions occur.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antioxidants/therapeutic use , Bleomycin/adverse effects , DNA Damage/drug effects , Lung/drug effects , Pregnatrienes/therapeutic use , Premedication , Animals , Antibiotics, Antineoplastic/metabolism , Antioxidants/metabolism , Bleomycin/metabolism , DNA Fragmentation/drug effects , Iron Chelating Agents/metabolism , Ligases/analysis , Lung/metabolism , Male , Poly Adenosine Diphosphate Ribose/metabolism , Pregnatrienes/metabolism , Rats , Rats, Sprague-Dawley
12.
Int J Clin Pharmacol Ther ; 35(1): 28-32, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9021439

ABSTRACT

The dose proportionality of tirilazad pharmacokinetics at dosages above 6.0 mg/kg/day were assessed in 18 healthy male volunteers between the ages of 19 and 46 years. Subjects were randomized to receive either 1.5 mg/kg, 3.0 mg/kg, or 4.0 mg/kg tirilazad mesylate every 6 hours for 29 doses (daily doses of 6.0, 12.0, and 16.0 mg/kg/day for 7 days). Each drug dose was administered intravenously over 10 minutes. Plasma tirilazad, U-89678, and U-87999 (active reduced metabolites) were quantified by HPLC. Two subjects in the high dose group withdrew before the end of the study. Following the first dose of tirilazad, dose-corrected pharmacokinetic parameters for all 3 compounds did not differ significantly among dose groups. After the final tirilazad the mean half-life of tirilazad was approximately 80 hours. Mean apparent tirilazad clearance did not differ significantly among groups. Mean U-89678 AUC0-6 following the last tirilazad dose did not differ significantly between the 6.0 and 12.0 mg/kg/day doses, but the value for the 16.0 mg/kg dose was higher than values from both lower doses (p = 0.044 and 0.056, respectively). Similar results were obtained for U-87999. The dose effects observed for the pharmacokinetics of these 2 metabolites may have been a function of intersubject variability. When combined with previous data concerning the dose proportionally of tirilazad pharmacokinetics at doses less than 6.0 mg/kg/day, the data from the present study suggest that the pharmacokinetics of tirilazad are approximately linear over a dosage range of 1.0-16.0 mg/kg/day. Due to the inability to assess the plasma protein binding of tirilazad and its reduced metabolites, the clinical significance of the departure from linearity of the pharmacokinetics of U-89678 and U-87999 cannot be directly assessed. Further study at higher doses will be needed to address this issue.


Subject(s)
Antioxidants/pharmacokinetics , Pregnatrienes/pharmacokinetics , Adult , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Male , Middle Aged , Oxidation-Reduction , Pregnatrienes/blood , Pregnatrienes/metabolism
13.
J Steroid Biochem Mol Biol ; 59(1): 49-54, 1996 Sep.
Article in English | MEDLINE | ID: mdl-9009237

ABSTRACT

RU 3117 belongs to a new series of steroids which exhibited a high relative binding affinity (RBA) for (+)[3H]PPP sites in rat testis membranes; its RBA was about 40 times higher than that of progesterone. Furthermore, it is devoid of any binding to classical steroid receptors; therefore in order to study its binding parameters on rat testis membranes it was tritiated. [3H]RU 3117 bound at least two distinct sites with Ka values of 0.4 +/- 0.06 x 10(9) M(-1) and 1.3 +/- 0.2 x 10(7) M(-1). Using this marker, competition studies with cold haloperidol showed that a part of this binding was haloperidol-sensitive, whereas another part was haloperidol-resistant. Interestingly, progesterone described as a sigma ligand competes with [3H]RU 3117 binding, with an RBA of 1.6%. When haloperidol was preincubated (250 nM) with rat testis membranes, in order to mask the sigma sites, we observed that DTG (1,3-di-O-tolylguanidine) and haloperidol displayed a very low RBA (< 0.1%) and were not able totally to displace the [3H]RU 3117 binding up to 50 microM. Furthermore, benztropine exhibited a significant RBA of 19% but its displacement curve showed a plateau (500-50,000 nM). These results showed that part of the haloperidol-resistant sites was benztropine sensitive but another part was displaced neither by haloperidol nor by benztropine. The presence of these remaining binding sites was confirmed by preincubating a mixture of haloperidol and benztropine with testis membranes. Under these conditions, [3H]RU 3117 displayed a Ka of 1.0 +/- 0.01 x 10(7) M(-1), and we observed that these sites were recognized, up to now, only by the steroids RU 1968 and RU 54173 which are also devoid of any binding to classical nuclear steroid receptors.


Subject(s)
Estrone/analogs & derivatives , Receptors, sigma/metabolism , Testis/metabolism , Animals , Benztropine/metabolism , Binding, Competitive , Estrenes/chemistry , Estrenes/metabolism , Estrone/chemistry , Estrone/metabolism , Haloperidol/metabolism , Kinetics , Male , Membranes/metabolism , Neurotransmitter Agents/metabolism , Pregnatrienes/chemistry , Pregnatrienes/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Steroids/metabolism
14.
Drug Metab Dispos ; 24(7): 697-701, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8818564

ABSTRACT

After administration of a mixed dose of both radioisotope and stable-isotope-labeled tirilazad, we carried out a parallel set of HPLC analyses for drug metabolites in bile samples from monkeys and dogs using either radioactivity monitoring (RAM) for 14C or the chemical reaction interface mass spectrometry technique (CRIMS) to detect 13C or 15N. CRIMS is a novel method where analytes are decomposed in a microwave-induced plasma and the elements contained in the analytes are reformulated into small gaseous species that are detected by a mass spectrometer. The comprehensiveness of detection, chromatographic resolution, sensitivity, signal/noise, and quantitative abilities of CRIMS were compared with RAM and in no case was RAM superior. This implies that stable isotopes may be substituted for radioisotopes in studies of drug metabolism where the ability of the latter approach to detect a label independent of the structures in which the label appears has been the primary reason for continuing to use a hazardous and expensive tracer. With HPLC-CRIMS, stable isotopes such as 13C and 15N can be comprehensively detected and quantitative patterns of drug metabolism from biological fluids can be produced that mirror the results when 14C is used.


Subject(s)
Bile/metabolism , Carbon Radioisotopes/chemistry , Free Radical Scavengers/analysis , Mass Spectrometry/methods , Pregnatrienes/analysis , Animals , Carbon Isotopes , Dogs , Macaca fascicularis , Male , Nitrogen Isotopes , Pregnatrienes/administration & dosage , Pregnatrienes/metabolism
15.
Chem Phys Lipids ; 76(1): 7-25, 1995 May 22.
Article in English | MEDLINE | ID: mdl-7788801

ABSTRACT

Oil-in-water emulsions are being used increasingly for the delivery of lipophilic drugs, but the fundamental physicochemical principles governing such delivery have not been explored. We determined the kinetics and thermodynamics of delivery from emulsions to cells in culture for two lipophilic compounds, U74006 and U74500. Two fundamental properties dominate the delivery, (a) the concentration of the compound in the lipid phase of the emulsion is directly proportional to the concentration of the compound in cells at equilibrium, and (b) the rate of transfer is directly proportional to the concentration of particles in contact with the cells. Thus, the transfer is consistent with direct partitioning from the lipid phase of the emulsion to cells and occurs by the direct collision of emulsion particles with cells. The details of the mechanism of delivery differ between the two compounds. Specifically, delivery of U74006 is first-order with respect to the drug accumulating in the cells. The transfer of U74500 is best described as a sum of two simultaneous pseudo first-order processes consistent with delivery from a single donor compartment to two receiver compartments. Furthermore, two molecules of U74500 appear to be involved in each transfer event. Our results show that relatively simple principles govern the delivery of compounds from oil-in-water emulsions to cells.


Subject(s)
Antioxidants/metabolism , Drug Delivery Systems , Pregnatrienes/metabolism , Animals , Antioxidants/pharmacology , Emulsions , Kinetics , Mice , Neuroblastoma/metabolism , Neurons/metabolism , Phosphatidylcholines/metabolism , Rats , Thermodynamics , Triolein/metabolism , Tumor Cells, Cultured
16.
Drug Metab Dispos ; 23(3): 383-92, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7628305

ABSTRACT

Tirilazad mesylate, a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage and head injury. In rat, tirilazad seems to be highly extracted and is cleared almost exclusively via hepatic elimination. The biotransformation of tirilazad has been investigated in liver microsomal preparations from adult male and female Sprague-Dawley rats. Tirilazad metabolism in male rat liver microsomes resulted in the formation of two primary metabolites: M1 and M2. In incubations with female rat liver microsomes, M2 was the only primary metabolite detected. Structural characterization of M1 and M2 by mass spectrometry demonstrated that M2 was formed by reduction of the delta 4-double bond in the steroid A-ring, whereas M1 arose from oxidative desaturation of one pyrrolidine ring. Further structural analysis of M2 by proton NMR demonstrated that reduction at C-5 had occurred by addition of hydrogen in the alpha-configuration. Using metabolic probes and antibodies specific to individual hepatic microsomal enzymes, CYP2C11 and 3-oxo-5 alpha-steroid:NADP+ delta 4-oxidoreductase (5 alpha-reductase) were identified as responsible for the formation of M1 and M2, respectively. The formation of M1 was inhibited by testosterone, nicotine, cimetidine, and anti-CYP2C11 IgG. The formation of M2 was inhibited by finasteride, a potent inhibitor of 5 alpha-reductase. Kinetic analysis of CYP2C11-mediated M1 formation in male rat liver microsomal incubations revealed that M1 formation occurred through a low-affinity/low-capacity process (KM = 16.67 microM, Vmax = 0.978 nmol/mg microsomal protein/min); the formation of M2 was mediated by 5 alpha-reductase in a high-affinity/low-capacity process (KM = 3.07 microM, Vmax = 1.06 nmol/mg microsomal protein/min). In contrast, the formation of M2 in female rat liver microsomes was mediated by 5 alpha-reductase in a high-affinity/high-capacity process (KM = 2.72 microM, Vmax = 4.11 nmol/mg microsomal protein/min). Comparison of calculated intrinsic formation clearances (Vmax/KM) for M1 and M2 indicated that the female rat possessed a greater in vitro metabolic capacity for tirilazad biotransformation than the male rat. Therefore, the clearance of tirilazad mesylate in the rat is mediated primarily by rat liver 5 alpha-reductase, and the capacity in the female rat is 5-fold the capacity in the male. These observations correlate with documented differences in 5 alpha-reductase activity and predict a gender difference in tirilazad hepatic clearance in vivo.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Lipid Peroxides/metabolism , Microsomes, Liver/metabolism , Pregnatrienes/metabolism , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism , Animals , Biotransformation , Cytochrome P450 Family 2 , Female , Gas Chromatography-Mass Spectrometry , Lipid Peroxides/pharmacokinetics , Magnetic Resonance Spectroscopy , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Molecular Structure , Pregnatrienes/pharmacokinetics , Protons , Rats , Rats, Sprague-Dawley
17.
Am J Physiol ; 267(6 Pt 2): H2371-7, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7810737

ABSTRACT

We tested whether, with exposure to an extraneous iron-catalyzed free radical-generating system, prevention of lipid peroxidation with U74006F, a 21-aminosteroid, could also prevent myocardial contractile dysfunction. Rabbits received either U74006F (10 mg/kg iv) or vehicle (V). Thirty minutes later the hearts were excised and perfused by a non-recirculating Langendorff technique. Six U74006F- and six V-treated hearts were exposed for 7.5 min to a .OH-generating system (H2O2 and Fe(2+)-ADP chelate). Myocardial lipid peroxides were measured by glutathione peroxidase-catalyzed oxidation of exogenous glutathione. With exposure to .OH, cytosolic lipid peroxide levels were increased threefold in V-treated hearts, but there was no increase in U74006F-treated hearts. After 30 min of recovery, developed pressure and maximum first derivative of left ventricular pressure were greater in U74006F-treated hearts than in V-treated hearts but were still 50 and 44% of levels in saline hearts, respectively. Coronary flow was markedly reduced after exposure to free radicals and was only slightly less depressed when U74006F was administered. When coronary flow following oxidant exposure was increased by nitroglycerin, U74006F again only modestly improved systolic function. Thus, although U74006F blocked lipid peroxidation, it only slightly improved the ventricular dysfunction caused by .OH. Therefore, factors other than lipid peroxidation play a major role in oxidant-induced myocardial stunning.


Subject(s)
Cardiomyopathies/prevention & control , Lipid Peroxidation/drug effects , Lipid Peroxides/antagonists & inhibitors , Myocardial Contraction/drug effects , Oxidative Stress , Pregnatrienes/therapeutic use , Adenosine Diphosphate/pharmacology , Animals , Cardiomyopathies/etiology , Coronary Circulation/drug effects , Diastole , Female , Ferrous Compounds/pharmacology , Free Radicals , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hydrogen Peroxide/pharmacology , L-Lactate Dehydrogenase/metabolism , Male , Myocardium/metabolism , Pregnatrienes/metabolism , Pregnatrienes/pharmacology , Rabbits , Systole
18.
J Crit Care ; 9(4): 213-22, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7889132

ABSTRACT

PURPOSE: The lazaroids are a new class of potent free-radical scavengers. We tested whether U-74389G, a lazaroid, could attenuate some of the adverse cardiopulmonary effects of sepsis. METHODS: Dogs were randomized to receive either 10 mg/kg U-74389G (n = 10), or a saline control (n = 11). After baseline measurements of hemodynamics and gas exchange, they were then randomized to receive either 0.2 mg/kg endotoxin or a saline infusion. Measurements of hemodynamics and gas exchange were repeated. The study was concluded 70 minutes after endotoxin infusion and the lungs were then removed for histologic evaluation. RESULTS: In endotoxin-treated control animals, PO2 decreased (278 +/- 123 mm Hg to 67 +/- 13 mm Hg, P < .05) and intrapulmonary shunt increased (12.9% +/- 1.1% to 28.2% +/- 11.4%, P < .05) after endotoxin. Pretreatment with U-74389G attenuated the decrease in PO2 (476 +/- 61 mm Hg to 226 +/- 143) and the increase in intrapulmonary shunt (12.6% +/- 6.1% to 14.3% +/- 6.8%) observed after endotoxin. The extent of lung injury and systemic hemodynamics were similar between control or U-74389G-treated dogs. CONCLUSIONS: A free-radical-scavenger can attenuate the gas exchange defect commonly associated with endotoxin but it does not improve the derangement of systemic hemodynamics.


Subject(s)
Antioxidants/metabolism , Endotoxins/adverse effects , Pregnatrienes/metabolism , Premedication , Pulmonary Gas Exchange/drug effects , Steroids, Heterocyclic/metabolism , Analysis of Variance , Animals , Blood Gas Analysis , Dogs , Endotoxins/metabolism , Free Radicals , Hemodynamics/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Pregnatrienes/therapeutic use , Pulmonary Edema , Random Allocation , Shock, Septic/metabolism , Steroids, Heterocyclic/therapeutic use , Vascular Resistance/drug effects
19.
J Pharmacol Exp Ther ; 269(1): 145-50, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8169818

ABSTRACT

Determination of the serum protein binding of tirilazad across species was required to predict the pharmacokinetic behavior of this new drug. Equilibrium dialysis and ultrafiltration techniques, commonly used to study serum protein binding, were shown to be unsuitable for tirilazad due to high nonspecific binding and low aqueous solubility, resulting in unbound drug levels that were nondetectable with current analytical methodology. Ultracentrifugation appeared to offer a technique with which unbound tirilazad could be measured; however, after extensive studies, the apparent lipid partitioning behavior of tirilazad into low density and very low density lipoproteins showed that ultracentrifugation was also unsuitable for determination of the true unbound fraction of tirilazad. Fractions of tirilazad measured in the supernatant were highly correlated with total triglycerides and very low density lipoproteins in the sera of all species analyzed. Studies with delipidized human serum yielded a nonsaturable binding isotherm with free fractions of less than 0.6 +/- 0.02% (mean +/- S.D.) over a concentration range of 4.6 to 55.6 micrograms/ml (normal human in vivo range, 0.01-20 micrograms/ml). These data indicated that, as the triglyceride content of the sera increases, portions of tirilazad bound to serum proteins shift into the lipid phase of lipoproteins. What effect this has on the true unbound fraction is unknown and does not seem to be ascertainable with current technology.


Subject(s)
Blood Proteins/metabolism , Dogs/blood , Macaca fascicularis/blood , Pregnatrienes/blood , Rats, Sprague-Dawley/blood , Acetonitriles/pharmacology , Adolescent , Adult , Aged , Animals , Binding, Competitive , Dialysis , Female , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Pregnatrienes/metabolism , Protein Binding , Rats , Species Specificity , Ultracentrifugation , Ultrafiltration
20.
J Steroid Biochem Mol Biol ; 44(3): 217-25, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8461255

ABSTRACT

The differential sensitivity of the rat liver glucocorticoid receptor (GR) to sulfhydryl group modifying agents when bound to various agonist and antagonist ligands was studied. [3H]Triamcinolone acetonide (TA) binding was completely abolished by previous treatment of the unbound receptor with various N-alkylmaleimides. On the contrary, [3H]RU486 binding was only slightly affected by treatment with N-ethylmaleimide (NEM) and more significantly decreased with maleimides bearing bulky substituents. Ligand exchange experiments demonstrated that, unlike the agonist TA, the antiglucocorticoid RU486 was unable to protect the GR binding site from the effect of NEM. This lack of protection would seem to be due to the presence of the bulky 11 beta-substituent in RU486 since RU26988 and RU28362, two 11 beta hydroxylated glucocorticoids bearing the same 17 alpha-propynyl side chain as RU486 but lacking the 11 beta-substituent could protect GR against NEM. The ability of a GR ligand to prevent NEM inactivation of TA binding appeared unrelated to its agonist or antagonist nature: deacylcortivazol, a potent agonist, afforded no protection whereas antagonists of the 17 beta-carboxamide series did. These data strongly suggest that compounds bearing bulky substituents on the steroid A and/or C rings, like deacylcortivazol and RU486, are positioned differently from canonical glucocorticoids in the steroid binding groove of the GR.


Subject(s)
Mifepristone/metabolism , Pregnatrienes/metabolism , Receptors, Glucocorticoid/metabolism , Sulfhydryl Reagents/pharmacology , Alkylation , Androstanols/metabolism , Animals , Binding, Competitive , Ethylmaleimide/pharmacology , Liver/metabolism , Male , Maleimides/pharmacology , Rats , Rats, Wistar , Receptors, Glucocorticoid/drug effects , Structure-Activity Relationship , Triamcinolone Acetonide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...