Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(11): e48385, 2012.
Article in English | MEDLINE | ID: mdl-23152771

ABSTRACT

Glucocorticoids (GCs) such as prednisolone are potent immunosuppressive drugs but suffer from severe adverse effects, including the induction of insulin resistance. Therefore, development of so-called Selective Glucocorticoid Receptor Modulators (SGRM) is highly desirable. Here we describe a non-steroidal Glucocorticoid Receptor (GR)-selective compound (Org 214007-0) with a binding affinity to GR similar to that of prednisolone. Structural modelling of the GR-Org 214007-0 binding site shows disturbance of the loop between helix 11 and helix 12 of GR, confirmed by partial recruitment of the TIF2-3 peptide. Using various cell lines and primary human cells, we show here that Org 214007-0 acts as a partial GC agonist, since it repressed inflammatory genes and was less effective in induction of metabolic genes. More importantly, in vivo studies in mice indicated that Org 214007-0 retained full efficacy in acute inflammation models as well as in a chronic collagen-induced arthritis (CIA) model. Gene expression profiling of muscle tissue derived from arthritic mice showed a partial activity of Org 214007-0 at an equi-efficacious dosage of prednisolone, with an increased ratio in repression versus induction of genes. Finally, in mice Org 214007-0 did not induce elevated fasting glucose nor the shift in glucose/glycogen balance in the liver seen with an equi-efficacious dose of prednisolone. All together, our data demonstrate that Org 214007-0 is a novel SGRMs with an improved therapeutic index compared to prednisolone. This class of SGRMs can contribute to effective anti-inflammatory therapy with a lower risk for metabolic side effects.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dibenzazepines/pharmacology , Receptors, Glucocorticoid/agonists , Thiadiazoles/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/genetics , Blood Glucose , Dibenzazepines/therapeutic use , Female , Gene Expression Regulation/drug effects , Humans , Kinetics , Liver/drug effects , Liver/enzymology , Male , Mice , Molecular Docking Simulation , Prednisolone/pharmacology , Prednisolone/therapeutic use , Protein Binding , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Thiadiazoles/therapeutic use
2.
J Clin Endocrinol Metab ; 97(5): E781-5, 2012 May.
Article in English | MEDLINE | ID: mdl-22419705

ABSTRACT

The TSH receptor (TSHR) on orbital fibroblasts (OF) is a proposed target of the autoimmune attack in Graves' ophthalmopathy. In the present study, we tested whether the novel low-molecular-weight (LMW) TSHR antagonist Org-274179-0 inhibits cAMP production induced by rhTSH, Graves' disease IgG (GD-IgG), or M22 (a potent human monoclonal TSHR stimulating antibody) in cultured and differentiated OF from Graves' ophthalmopathy patients. cAMP production significantly increased after incubation either with 10 mU/ml rhTSH (3-fold; P ≤ 0.05), 1 mg/ml GD-IgG (2-fold; P ≤ 0.05), or 500 ng/ml M22 (5-fold; P ≤ 0.05). Incubation with the LMW TSHR antagonist dose dependently inhibited rhTSH, GD-IgG as well as the M22-induced cAMP production at nanomolar concentrations; complete blockade was affected at 10(-6) M. Our results suggest that GD-IgG- and M22-induced cAMP production in differentiated OF is exclusively mediated via the TSHR because it can be completely blocked by the LMW TSHR antagonist, Org 274179-0.


Subject(s)
Aminoquinolines/pharmacology , Antibodies, Monoclonal/pharmacology , Cyclic AMP/biosynthesis , Fibroblasts/drug effects , Graves Disease/immunology , Immunoglobulin G/pharmacology , Receptors, Thyrotropin/antagonists & inhibitors , Thyrotropin/pharmacology , Animals , CHO Cells , Cricetinae , Fibroblasts/metabolism , Humans , Orbit/cytology
3.
Br J Pharmacol ; 165(7): 2314-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22014107

ABSTRACT

BACKGROUND AND PURPOSE: Graves' disease (GD) is an autoimmune disease in which the thyroid is overactive, producing excessive amounts of thyroid hormones, caused by thyroid-stimulating hormone (TSH) receptor-stimulating immunoglobulins (TSIs). Many GD patients also suffer from thyroid eye disease (Graves' ophthalmopathy or GO), as TSIs also activate TSH receptors in orbital tissue. We recently developed low molecular weight (LMW) TSH receptor antagonists as a novel therapeutic strategy for the treatment of GD and GO. Here, we determined the molecular pharmacology of a prototypic, nanomolar potent LMW TSH receptor antagonist, Org 274179-0. EXPERIMENTAL APPROACH: Using CHO cells heterogeneously expressing human TSH receptors and rat FRTL-5 cells endogenously expressing rat TSH receptors, we determined the potency and efficacy of Org 274179-0 at antagonizing TSH- and TSI-induced TSH receptor signalling and its cross-reactivity at related follicle-stimulating hormone and luteinizing hormone receptors. We analysed the allosteric mode of interaction of Org 274179-0 and determined whether it is an inverse agonist at five naturally occurring, constitutively active TSH receptor mutants. KEY RESULTS: Nanomolar concentrations of Org 274179-0 completely inhibited TSH (and TSI)-mediated TSH receptor activation with little effect on the potency of TSH, in accordance with an allosteric mechanism of action. Conversely, increasing levels of TSH receptor stimulation only marginally reduced the antagonist potency of Org 274179-0. Org 274179-0 fully blocked the increased basal activity of all the constitutively active TSH receptor mutants tested with nanomolar potencies. CONCLUSIONS AND IMPLICATIONS: Nanomolar potent TSH receptor antagonists like Org 274179-0 have therapeutic potential for the treatment of GD and GO.


Subject(s)
Aminoquinolines/pharmacology , Receptors, Thyrotropin/antagonists & inhibitors , Adenylyl Cyclases/metabolism , Aminoquinolines/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Evaluation, Preclinical , Graves Disease/drug therapy , Graves Disease/immunology , Humans , Molecular Weight , Mutagenesis, Site-Directed , Rats , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Signal Transduction/drug effects , Thyrotropin/metabolism
4.
J Comput Chem ; 32(11): 2441-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21598279

ABSTRACT

The biosynthesis of the mineralocorticoid hormone aldosterone involves a multistep hydroxylation of 11-deoxycorticosterone at the 11- and 18-positions, resulting in the formation of corticosterone and 18-hydroxycorticosterone, the final precursor of aldosterone. Two members of the cytochrome P450 11B family, CYP11B1 and CYP11B2, are known to catalyze these 11- and 18-hydroxylations, however, only CYP11B2 can oxidize 18-hydroxycorticosterone to aldosterone. It is unknown what sequence of hydroxylations leads to the formation of 18-hydroxycorticosterone. In this study we have investigated which of the possible conversion paths towards formation of 18-hydroxycorticosterone and aldosterone are most likely from the ligand perspective. Therefore, we combined quantum mechanical investigations on the steroid conformations of 11-deoxycorticosterone and its ensuing reaction intermediates with Fukui indices calculations to predict the reactivity of their carbon atoms for an attack by the iron-oxygen species. Both F(-) and F(0) were calculated to account for different mechanisms of substrate conversion. We show which particular initial conformations of 11-deoxycorticosterone and which conversion paths are likely to result in the successful synthesis of aldosterone, and thereby may be representative for the mechanism of aldosterone biosynthesis by CYP11B2. Moreover, we found that the most likely path for aldosterone synthesis coincides with the substrate conformation proposed in an earlier publication. To summarize, we show that on a theoretical and strictly ligand-directed basis only a limited number of reaction paths in the conversion of 11-deoxycorticosterone to aldosterone is possible. Despite its theoretical nature, this knowledge may help to understand the catalytic function of CYP11B1 and CYP11B2.


Subject(s)
Aldosterone/biosynthesis , Aldosterone/chemistry , Cytochrome P-450 CYP11B2/chemistry , Ligands , Quantum Theory , Iron/chemistry , Molecular Structure , Oxygen/chemistry
5.
J Med Chem ; 53(4): 1712-25, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20121113

ABSTRACT

Reducing aldosterone action is beneficial in various major diseases such as heart failure. Currently, this is achieved with mineralocorticoid receptor antagonists, however, aldosterone synthase (CYP11B2) inhibitors may offer a promising alternative. In this study, we used three-dimensional modeling of CYP11B2 to model the binding modes of the natural substrate 18-hydroxycorticosterone and the recently published CYP11B2 inhibitor R-fadrozole as a rational guide to design 44 structurally simple and achiral 1-benzyl-1H-imidazoles. Their syntheses, in vitro inhibitor potencies, and in silico docking are described. Some promising CYP11B2 inhibitors were identified, with our novel lead MOERAS115 (4-((5-phenyl-1H-imidazol-1-yl)methyl)benzonitrile) displaying an IC(50) for CYP11B2 of 1.7 nM, and a CYP11B2 (versus CYP11B1) selectivity of 16.5, comparable to R-fadrozole (IC(50) for CYP11B2 6.0 nM, selectivity 19.8). Molecular docking of the inhibitors in the models enabled us to generate posthoc hypotheses on their binding modes, providing a valuable basis for future studies and further design of CYP11B2 inhibitors.


Subject(s)
Benzyl Compounds/chemical synthesis , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Imidazoles/chemical synthesis , Models, Molecular , 18-Hydroxycorticosterone/chemistry , Animals , Benzyl Compounds/chemistry , Benzyl Compounds/pharmacology , Catalytic Domain , Cell Line , Cricetinae , Cricetulus , Cytochrome P-450 CYP11B2/chemistry , Fadrozole/chemistry , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Molecular Dynamics Simulation , Protein Binding , Stereoisomerism , Structure-Activity Relationship
6.
Endocrinology ; 149(1): 28-31, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17884944

ABSTRACT

Reversal of cardiac fibrosis is a major determinant of the salutary effects of mineralocorticoid receptor antagonists in heart failure. Recently, R-fadrozole was coined as an aldosterone biosynthesis inhibitor, offering an appealing alternative to mineralocorticoid receptor antagonists to block aldosterone action. The present study aimed to evaluate the effects of R- and S-fadrozole on plasma aldosterone and urinary aldosterone excretion rate and to compare their effectiveness vs. the mineralocorticoid receptor antagonist potassium canrenoate to reverse established cardiac fibrosis. Male lean spontaneously hypertensive heart failure (SHHF) rats (40 wk) were treated for 8 wk by sc infusions of low (0.24 mg/kg.d) or high (1.2 mg/kg.d) doses of R- or S-fadrozole or by potassium canrenoate via drinking water (7.5 mg/kg.d). At the high dose, plasma aldosterone levels were decreased similarly by R- and S-fadrozole, whereas urinary aldosterone excretion rate was reduced only by S-fadrozole. In contrast, whereas at the high dose, R-fadrozole effectively reversed preexistent left ventricular interstitial fibrosis by 50% (vs. 42% for canrenoate), S-fadrozole was devoid of an antifibrotic effect. The low doses of the fadrozole enantiomers did not change cardiac fibrosis or plasma aldosterone but similarly reduced urinary aldosterone excretion rate. In conclusion, R-fadrozole may possess considerable therapeutic merit because of its potent antifibrotic actions in the heart. However, the observed discordance between the aldosterone-lowering and antifibrotic effects of the fadrozole enantiomers raises some doubt about the mechanism by which R-fadrozole diminishes cardiac collagen and about the generality of the concept of lowering aldosterone levels to treat the diseased heart.


Subject(s)
Aldosterone/blood , Fadrozole/chemistry , Fadrozole/therapeutic use , Heart Failure/prevention & control , Heart/drug effects , Myocardium/pathology , Aldosterone/urine , Animals , Canrenoic Acid/pharmacology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/genetics , Collagen Type III/metabolism , Drug Evaluation, Preclinical , Fibrosis , Gene Expression Regulation/drug effects , Heart Failure/urine , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Myocardium/metabolism , Rats , Rats, Inbred SHR , Stereoisomerism , Structure-Activity Relationship , Treatment Outcome
7.
J Comput Aided Mol Des ; 21(8): 455-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17646925

ABSTRACT

Aldosterone is synthesised by aldosterone synthase (CYP11B2). CYP11B2 has a highly homologous isoform, steroid 11beta-hydroxylase (CYP11B1), which is responsible for the biosynthesis of aldosterone precursors and glucocorticoids. To investigate aldosterone biosynthesis and facilitate the search for selective CYP11B2 inhibitors, we constructed three-dimensional models for CYP11B1 and CYP11B2 for both human and rat. The models were constructed based on the crystal structure of Pseudomonas Putida CYP101 and Oryctolagus Cuniculus CYP2C5. Small steric active site differences between the isoforms were found to be the most important determinants for the regioselective steroid synthesis. A possible explanation for these steric differences for the selective synthesis of aldosterone by CYP11B2 is presented. The activities of the known CYP11B inhibitors metyrapone, R-etomidate, R-fadrazole and S-fadrazole were determined using assays of V79MZ cells that express human CYP11B1 and CYP11B2, respectively. By investigating the inhibitors in the human CYP11B models using molecular docking and molecular dynamics simulations we were able to predict a similar trend in potency for the inhibitors as found in the in vitro assays. Importantly, based on the docking and dynamics simulations it is possible to understand the enantioselectivity of the human enzymes for the inhibitor fadrazole, the R-enantiomer being selective for CYP11B2 and the S-enantiomer being selective for CYP11B1.


Subject(s)
Computer Simulation , Cytochrome P-450 CYP11B2/chemistry , Steroid 11-beta-Hydroxylase/chemistry , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Drug Design , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Rats , Sequence Homology, Amino Acid , Steroid 11-beta-Hydroxylase/antagonists & inhibitors , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Thermodynamics
8.
Bioorg Med Chem ; 10(4): 1143-52, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11836126

ABSTRACT

A series of 1,3-diazacycloalkyl carboxaldehyde oxime derivatives was synthesized and tested for muscarinic activity in receptor binding assays using [3H]-oxotremorine-M (OXO-M) and [3H]-pirenzepine (PZ) as ligands. Potential muscarinic agonistic or antagonistic properties of the compounds were determined using binding studies measuring their potencies to inhibit the binding of OXO-M and PZ. Preferential inhibition of OXO-M binding was used as an indicator for potential muscarinic agonistic properties; this potential was confirmed in functional studies on isolated organs.


Subject(s)
Cholinergic Agents/chemical synthesis , Oximes/pharmacology , Animals , Arecoline/chemistry , Arecoline/pharmacology , Binding, Competitive , Cholinergic Agents/pharmacology , Heart Atria/drug effects , Ileum/drug effects , Muscarinic Agonists/chemical synthesis , Muscarinic Agonists/pharmacology , Oximes/chemical synthesis , Protein Binding , Radioligand Assay , Rats , Receptors, Muscarinic/metabolism , Structure-Activity Relationship , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...