Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Front Physiol ; 13: 923091, 2022.
Article in English | MEDLINE | ID: mdl-35910566

ABSTRACT

Introduction: Rewarming from accidental hypothermia is often complicated by hypothermia-induced cardiovascular dysfunction, which could lead to shock. Current guidelines do not recommend any pharmacological treatment at core temperatures below 30°C, due to lack of knowledge. However, previous in vivo studies have shown promising results when using phosphodiesterase 3 (PDE3) inhibitors, which possess the combined effects of supporting cardiac function and alleviating the peripheral vascular resistance through changes in cyclic nucleotide levels. This study therefore aims to investigate whether PDE3 inhibitors milrinone, amrinone, and levosimendan are able to modulate cyclic nucleotide regulation in hypothermic settings. Materials and methods: The effect of PDE3 inhibitors were studied by using recombinant phosphodiesterase enzymes and inverted erythrocyte membranes at six different temperatures-37°C, 34°C, 32°C, 28°C, 24°C, and 20°C- in order to evaluate the degree of enzymatic degradation, as well as measuring cellular efflux of both cAMP and cGMP. The resulting dose-response curves at every temperature were used to calculate IC50 and Ki values. Results: Milrinone IC50 and Ki values for cGMP efflux were significantly lower at 24°C (IC50: 8.62 ± 2.69 µM) and 20°C (IC50: 7.35 ± 3.51 µM), compared to 37°C (IC50: 22.84 ± 1.52 µM). There were no significant changes in IC50 and Ki values for enzymatic breakdown of cAMP and cGMP. Conclusion: Milrinone, amrinone and levosimendan, were all able to suppress enzymatic degradation and inhibit extrusion of cGMP and cAMP below 30°C. Our results show that these drugs have preserved effect on their target molecules during hypothermia, indicating that they could provide an important treatment option for hypothermia-induced cardiac dysfunction.

3.
Front Physiol ; 12: 695779, 2021.
Article in English | MEDLINE | ID: mdl-34393818

ABSTRACT

INTRODUCTION: Cardiovascular dysfunction is a potentially lethal complication of hypothermia. Due to a knowledge gap, pharmacological interventions are not recommended at core temperatures below 30°C. Yet, further cooling is induced in surgical procedures and survival of accidental hypothermia is reported after rewarming from below 15°C, advocating a need for evidence-based treatment guidelines. In vivo studies have proposed vasodilation and afterload reduction through arteriole smooth muscle cGMP-elevation as a favorable strategy to prevent cardiovascular dysfunction in hypothermia. Further development of treatment guidelines demand information about temperature-dependent changes in pharmacological effects of clinically relevant vasodilators. MATERIALS AND METHODS: Human phosphodiesterase-enzymes and inverted erythrocytes were utilized to evaluate how vasodilators sildenafil and vardenafil affected cellular efflux and enzymatic breakdown of cAMP and cGMP, at 37°C, 34°C, 32°C, 28°C, 24°C, and 20°C. The ability of both drugs to reach their cytosolic site of action was assessed at the same temperatures. IC50- and K i -values were calculated from dose-response curves at all temperatures, to evaluate temperature-dependent effects of both drugs. RESULTS: Both drugs were able to reach the intracellular space at all hypothermic temperatures, with no reduction compared to normothermia. Sildenafil IC50 and K i -values increased during hypothermia for enzymatic breakdown of both cAMP (IC50: 122 ± 18.9 µM at 37°C vs. 269 ± 14.7 µM at 20°C, p < 0.05) and cGMP (IC50: 0.009 ± 0.000 µM at 37°C vs. 0.024 ± 0.004 µM at 32°C, p < 0.05), while no significant changes were detected for vardenafil. Neither of the drugs showed significant hypothermia-induced changes in IC50 and K i- values for inhibition of cellular cAMP and cGMP efflux. CONCLUSION: Sildenafil and particularly vardenafil were ableto inhibit elimination of cGMP down to 20°C. As the cellular effects of these drugs can cause afterload reduction, they show potential in treating cardiovascular dysfunction during hypothermia. As in normothermia, both drugs showed higher selectivity for inhibition of cGMP-elimination than cAMP-elimination at low core temperatures, indicating that risk for cardiotoxic side effects is not increased by hypothermia.

4.
J Steroid Biochem Mol Biol ; 213: 105951, 2021 10.
Article in English | MEDLINE | ID: mdl-34271023

ABSTRACT

The biodynamics and biokinetics of sex hormones are complex. In addition to the classical steroid receptors (nuclear receptors), these hormones act through several non-genomic mechanisms. Modulation of ABC-transporters by progesterone represents a non-genomic mechanism. In the present study, we employed inside out vesicles from human erythrocytes to characterize high affinity cGMP transport by ABCC5 (member 5 of the ATP-Binding Cassette subfamily C). Progesterone and testosterone inhibited the transport with respective Ki of 1.2 ± 0.3 and 2.0 ± 0.6 µmol/L. We used virtual ligand screening (VLS) to identify analogues to progesterone and testosterone. A large number of substances were screened in silico and the 19 most promising candidates were screened in vitro. Each substance was tested for a concentration of 10 µmol/L. The range of cGMP transport reduction was 21.5% to 86.2% for progesterone analogues and 8.6% to 93.8 % for testosterone analogues. Three of the most potent test compounds (TC) of each analogue class, in addition to progesterone and testosterone, were characterized for concentrations from 1 nanomol/L to 1 mmol/L. The progesterone analogues showed following Ki-values (µmol/L): TC-08: 0.61, TC-16: 0.66 and TC-15: 9.3. The Ki-values (µmol/L) for the testosterone analogues were: TC-18: 0.10, TC-07: 0.67 andTC-05: 2.0. The present study shows that VLS may be a versatile tool in the development of membrane transport modulating agents (MTMAs).


Subject(s)
Cyclic GMP/metabolism , Erythrocyte Membrane/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Progesterone/pharmacology , Testosterone/pharmacology , Biological Transport/drug effects , Dose-Response Relationship, Drug , Erythrocyte Membrane/metabolism , Gene Expression , High-Throughput Screening Assays , Humans , Kinetics , Ligands , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/genetics , Progesterone/analogs & derivatives , Protein Binding , Structure-Activity Relationship , Testosterone/analogs & derivatives , User-Computer Interface
5.
Bioorg Med Chem ; 36: 116060, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33691270

ABSTRACT

The dominant sex hormone testosterone is mainly metabolized by liver enzymes belonging to the uridine-diphospho (UDP) glucuronosyltransferase (UGT) family. These enzymes are the main phase II enzymes, and they have an important role in the detoxification of endogenous and exogenous compounds in humans. The aim of the present study was to improve the understanding of the binding properties of UGT2B17. A homology modelling procedure was used to generate models of the UGT2B17 enzyme based on templates with known crystal structures. Molecular docking of inhibitors was performed to gain further insights in the interactions between ligand and binding site, and to determine which of the models had the best accuracy. ROC curves were made to evaluate the ability of the models to differentiate between binders (inhibitors) and non-binders (decoys). When comparing the four models, which were based on four different crystal structures, the model based on the 4AMG crystal structure was the most accurate in distinguishing between true binders and non-binders. Investigating pharmacological UGT2B17 inhibition may provide novel treatment for patients with low testosterone levels. Such treatment may elevate endogenous testosterone levels and provide a more predictable increase in serum concentrations rather than un-physiological elevation of serum levels through direct treatment with testosterone, and this could be favorable both for giving a predictable treatment regime with reduced chances of serious adverse effects. The present study may serve as a tool in the search for novel drugs aiming for increasing testosterone levels.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucuronosyltransferase/antagonists & inhibitors , Testosterone/pharmacology , Enzyme Inhibitors/chemistry , Glucuronosyltransferase/metabolism , Humans , Minor Histocompatibility Antigens/metabolism , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Testosterone/chemistry
6.
Biomed Pharmacother ; 126: 110109, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32229414

ABSTRACT

BACKGROUND: Clinical studies have reported overexpression of PDE5 and elevation of intracellular cyclic GMP in various types of cancer cells. ABCC5 transports cGMP out of the cells with high affinity. PDE5 inhibitors prevent both cellular metabolism and cGMP efflux by inhibiting ABCC5 as well as PDE5. Increasing intracellular cGMP is hypothesized to promote apoptosis and growth restriction in tumor cells and also has potential for clinical use in treatment of cardiovascular disease and erectile dysfunction. Vardenafil is a potent inhibitor of both PDE5 and ABCC5-mediated cGMP cellular efflux. Nineteen novel vardenafil analogs that have been predicted as potent inhibitors by VLS were chosen for tests of their ability to inhibit ATP- dependent transport of cGMP by measuring the accumulation of cyclic GMP in inside-out vesicles. AIM: In this study, we investigated the ability of nineteen new compounds to inhibit ABCC5- mediated cGMP transport. We also determined the Ki values of the six most potent compounds. METHODS: Preparation of human erythrocyte inside out vesicles and transport assay. RESULTS: Ki values for six of nineteen compounds that showed more than 50 % inhibition of cGMP transport in the screening test were determined and ranged from 1.1 to 23.1 µM. One compound was significantly more potent than the positive control, sildenafil. CONCLUSION: Our findings show that computational screening correctly identified vardenafil-analogues that potently inhibit cGMP efflux-pumps from cytosol and could have substantial clinical potential in treatment of patients with diverse disorders.


Subject(s)
Cyclic GMP/metabolism , Drug Discovery , Molecular Docking Simulation , Molecular Dynamics Simulation , Vardenafil Dihydrochloride/chemistry , Vardenafil Dihydrochloride/pharmacology , Binding Sites , Biological Transport/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Molecular Conformation , Phosphodiesterase 5 Inhibitors/pharmacology , Protein Binding , Structure-Activity Relationship
7.
Future Med Chem ; 10(2): 135-155, 2018 01.
Article in English | MEDLINE | ID: mdl-29235904

ABSTRACT

AIM: Low oxytocin (OT) level is involved in a number of psychiatric diseases, indicating that OT could be used to aid treating these disorders. OT itself is unable to cross the blood-brain barrier, and development of new small nonpeptide drugs targeting the OT receptor (OXTR) may be beneficial for treating mental disorders. Results & methodology: Three OXTR models were constructed based on crystallized homologous proteins (Protein Data Bank [PDB]: 2Y00, PDB: 4BVN and PDB: 4LDE). The abilities of the models to discriminate between true binders and decoys were analyzed using receiver operating characteristics curves, and the 4LDE-based model gave the best result. CONCLUSION: The present study demonstrates that the 4LDE-based model may be suitable as a tool for the development of novel drugs targeting OXTR.


Subject(s)
Molecular Docking Simulation , Receptors, Oxytocin/chemistry , Amino Acid Sequence , Humans , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Sequence Alignment
8.
J Pharm Pharmacol ; 69(6): 675-683, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28211580

ABSTRACT

OBJECTIVES: To determine the ability of 11 sildenafil analogues to discriminate between cyclic nucleotide phosphodiesterases (cnPDEs) and to characterise their inhibitory potencies (Ki values) of PDE5A1-dependent guanosine cyclic monophosphate (cGMP) hydrolysis. METHODS: Sildenafil analogues were identified by virtual ligand screening (VLS) and screened for their ability to inhibit adenosine cyclic monophosphate (cAMP) hydrolysis by PDE1A1, PDE1B1, PDE2A1, PDE3A, PDE10A1 and PDE10A2, and cGMP hydrolysis by PDE5A, PDE6C, PDE9A2 for a low (1 nm) and high concentration (10 µm). Complete IC50 plots for all analogues were performed for PDE5A-dependent cGMP hydrolysis. Docking studies and scoring were made using the ICM molecular modelling software. KEY FINDINGS: The analogues in a low concentration showed no or low inhibition of PDE1A1, PDE1B1, PDE2A1, PDE3A, PDE10A1 and PDE10A2. In contrast, PDE5A and PDE6C were markedly inhibited to a similar extent by the analogues in a low concentration, whereas PDE9A2 was much less inhibited. The analogues showed a relative narrow range of Ki values for PDE5A inhibition (1.2-14 nm). The sildenafil molecule was docked in the structure of PDE5A1 co-crystallised with sildenafil. All the analogues had similar binding poses as sildenafil. CONCLUSIONS: Sildenafil analogues that inhibit cellular cGMP efflux are potent inhibitors of PDE5A and PDE6C.


Subject(s)
Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Phosphodiesterase Inhibitors/pharmacology , Sildenafil Citrate/pharmacology , Cyclic AMP/metabolism , Humans , Hydrolysis/drug effects
9.
Eur J Med Chem ; 94: 229-36, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25768705

ABSTRACT

Herein, we describe the synthesis, biological evaluation and molecular docking of the selective PPARß/δ antagonist (4-methyl-2-(4-(trifluoromethyl)phenyl)-N-(2-(5-(trifluoromethyl)-pyridin-2-ylsulfonyl)ethyl)thiazole-5-carboxamide)), CC618. Results from in vitro luciferase reporter gene assays against the three known human PPAR subtypes revealed that CC618 selectively antagonizes agonist-induced PPARß/δ activity with an IC50 = 10.0 µM. As observed by LC-MS/MS analysis of tryptic digests, the treatment of PPARß/δ with CC618 leads to a covalent modification of Cys249, located centrally in the PPARß/δ ligand binding pocket, corresponding to the conversion of its thiol moiety to a 5-trifluoromethyl-2-pyridylthioether. Finally, molecular docking is employed to shed light on the mode of action of the antagonist and its structural consequences for the PPARß/δ ligand binding pocket.


Subject(s)
Models, Molecular , PPAR delta/antagonists & inhibitors , PPAR-beta/antagonists & inhibitors , Sulfones/chemistry , Sulfones/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , Sulfones/chemical synthesis , Thiazoles/chemical synthesis
10.
Eur J Pharmacol ; 745: 249-53, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25445042

ABSTRACT

Intracellular cyclic nucleotides are eliminated by phosphodiesterases (PDEs) and by ATP Binding cassette transporters such as ABCC4 and ABCC5. PDE5 and ABCC5 have similar affinity for cGMP whereas ABCC5 has much higher affinity for cGMP compared with cAMP. Since the substrate (cGMP) is identical for these two eliminatory processes it is conceivable that various PDE inhibitors also modulate ABCC5-transport. Cyclic GMP is also transported by ABBC4 but the affinity is much lower with a Km 50-100 times higher than for that of ABBCC5. The present study aimed to determine Ki-values for specific or relative specific PDE5 inhibitors (vardenafil, tadalafil, zaprinast and dipyridamole) and the non-specific PDE inhibitors (IBMX, caffeine and theophylline) for ABCC5 and ABCC4 transport. The transport of [(3)H]-cGMP (2 µM) was concentration-dependently inhibited with the following Ki-values: vardenafil (0.62 µM), tadalafil (14.1 µM), zaprinast (0.68 µM) and dipyridamole (1.2 µM), IBMX (10 µM), caffeine (48 µM) and theophylline (69 µM). The Ki-values for the inhibition of the [(3)H]-cAMP (2 µM) transport were: vardenafil (3.4 µM), tadalafil (194 µM), zaprinast (2.8 µM), dipyridamole (5.5 µM), IBMX (16 µM), caffeine (41 µM) and theophylline (85 µM). The specificity for ABCC5 we defined as ratio between Ki-values for inhibition of [(3)H]-cGMP and [(3)H]-cAMP transport. Tadalafil showed the highest specificity (Ki-ratio: 0.073) and caffeine the lowest (Ki-ratio: 1.2).


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Caffeine/pharmacology , Carbolines/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dipyridamole/pharmacology , Humans , Imidazoles/pharmacology , Kinetics , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Purinones/pharmacology , Sulfones/pharmacology , Tadalafil , Theophylline/pharmacology , Triazines/pharmacology , Vardenafil Dihydrochloride
11.
Int J Mol Sci ; 15(3): 4878-902, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24651460

ABSTRACT

The mitogen-activated protein kinase-activated protein kinase MK5 is a substrate of the mitogen-activated protein kinases p38, ERK3 and ERK4. Cell culture and animal studies have demonstrated that MK5 is involved in tumour suppression and promotion, embryogenesis, anxiety, cell motility and cell cycle regulation. In the present study, homology models of MK5 were used for molecular dynamics (MD) simulations of: (1) MK5 alone; (2) MK5 in complex with an inhibitor; and (3) MK5 in complex with the interaction partner p38α. The calculations showed that the inhibitor occupied the active site and disrupted the intramolecular network of amino acids. However, intramolecular interactions consistent with an inactive protein kinase fold were not formed. MD with p38α showed that not only the p38 docking region, but also amino acids in the activation segment, αH helix, P-loop, regulatory phosphorylation region and the C-terminal of MK5 may be involved in forming a very stable MK5-p38α complex, and that p38α binding decreases the residual fluctuation of the MK5 model. Electrostatic Potential Surface (EPS) calculations of MK5 and p38α showed that electrostatic interactions are important for recognition and binding.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Molecular Dynamics Simulation , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 14/chemistry , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinase Kinases/chemistry , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Static Electricity , Substrate Specificity , Thermodynamics
12.
J Chem Inf Model ; 54(3): 933-43, 2014 Mar 24.
Article in English | MEDLINE | ID: mdl-24521202

ABSTRACT

The serotonin (5-hydroxytryptamine, 5-HT) transporter (SERT) plays an essential role in the termination of serotonergic neurotransmission by removing 5-HT from the synaptic cleft into the presynaptic neuron. It is also of pharmacological importance being targeted by antidepressants and psychostimulant drugs. Here, five commercial databases containing approximately 3.24 million drug-like compounds have been screened using a combination of two-dimensional (2D) fingerprint-based and three-dimensional (3D) pharmacophore-based screening and flexible docking into multiple conformations of the binding pocket detected in an outward-open SERT homology model. Following virtual screening (VS), selected compounds were evaluated using in vitro screening and full binding assays and an in silico hit-to-lead (H2L) screening was performed to obtain analogues of the identified compounds. Using this multistep VS/H2L approach, 74 active compounds, 46 of which had K(i) values of ≤1000 nM, belonging to 16 structural classes, have been identified, and multiple compounds share no structural resemblance with known SERT binders.


Subject(s)
Psychotropic Drugs/chemistry , Psychotropic Drugs/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Binding Sites , Databases, Pharmaceutical , Drug Discovery , Humans , Molecular Docking Simulation , Protein Binding , Serotonin Plasma Membrane Transport Proteins/chemistry
13.
Theor Biol Med Model ; 10: 56, 2013 Sep 14.
Article in English | MEDLINE | ID: mdl-24034446

ABSTRACT

BACKGROUND: Mitogen-activated protein kinase-activated protein kinase 5 (MK5) is involved in one of the major signaling pathways in cells, the mitogen-activated protein kinase pathway. MK5 was discovered in 1998 by the groups of Houng Ni and Ligou New, and was found to be highly conserved throughout the vertebrates. Studies, both in vivo and in vitro, have shown that it is implicated in tumor suppression as well as tumor promotion, embryogenesis, anxiety, locomotion, cell motility and cell cycle regulation. METHODS: In order to obtain a molecular model of MK5 that can be used as a working tool for development of chemical probes, three MK5 models were constructed and refined based on three different known crystal structures of the closely related MKs; MK2 [PDB: 2OZA and PDB: 3M2W] and MK3 [PDB: 3FHR]. The main purpose of the present MK5 molecular modeling study was to identify the best suited template for making a MK5 model. The ability of the generated models to effectively discriminate between known inhibitors and decoys was analyzed using receiver operating characteristic (ROC) curves. RESULTS: According to the ROC curve analyzes, the refined model based on 3FHR was most effective in discrimination between known inhibitors and decoys. CONCLUSIONS: The 3FHR-based MK5 model may serve as a working tool for development of chemical probes using computer aided drug design. The biological function of MK5 still remains elusive, but its role as a possible drug target may be elucidated in the near future.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Docking Simulation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Structural Homology, Protein , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Ligands , Molecular Sequence Data , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , ROC Curve , Static Electricity
14.
Chem Biol Drug Des ; 81(6): 695-706, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23574807

ABSTRACT

Twelve alkyl analogues (1-12) of the high-affinity serotonin transporter (SERT) inhibitor 6-nitroquipazine (6-NQ) were synthesized and studied using in vitro radioligand competition binding assays to determine their binding affinity (Ki ). The putative antidepressant activity of five of the binders with the highest SERT binding affinities was studied by the forced swim and locomotor activity mouse tests. The three-dimensional (3D) structures of 8 and 9 were determined using NOE NMR technique. Flexible docking of the compounds was undertaken to illustrate the binding of the compounds in the SERT model. Our results showed that several of the 6-NQ analogues are high-affinity SERT inhibitors and indicated that the octyl (8), decyl (10) and dodecyl (12) 6-NQ analogues exhibit moderate antidepressant activity.


Subject(s)
Antidepressive Agents/chemical synthesis , Quipazine/analogs & derivatives , Selective Serotonin Reuptake Inhibitors/chemical synthesis , Serotonin Plasma Membrane Transport Proteins/chemistry , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Binding Sites , Male , Mice , Molecular Docking Simulation , Motor Activity/drug effects , Protein Binding , Protein Structure, Tertiary , Quipazine/chemical synthesis , Quipazine/chemistry , Quipazine/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology
15.
J Med Chem ; 55(7): 3049-57, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22380603

ABSTRACT

Elevated intracellular levels of cyclic guanosine monophosphate (cGMP) may induce apoptosis, and at least some cancer cells seem to escape this effect by increased efflux of cGMP, as clinical studies have shown that extracellular cGMP levels are elevated in various types of cancer. The human ATP binding cassette (ABC) transporter ABCC5 transports cGMP out of cells, and inhibition of ABCC5 may have cytotoxic effects. Sildenafil inhibits cGMP efflux by binding to ABCC5, and in order to search for potential novel ABCC5 inhibitors, we have identified sildenafil derivates using structural and computational guidance and tested them for the cGMP efflux effect. Eleven compounds from virtual ligand screening (VLS) were tested in vitro, using inside-out vesicles (IOV), for inhibition of cGMP efflux. Seven of 11 compounds predicted by VLS to bind to ABCC5 were more potent than sildenafil, and the two most potent showed K(i) of 50-100 nM.


Subject(s)
Cyclic GMP/antagonists & inhibitors , Databases, Factual , Models, Molecular , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Piperazines/chemistry , Sulfones/chemistry , Amino Acid Sequence , Animals , Cyclic GMP/metabolism , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/ultrastructure , Humans , Ligands , Mice , Molecular Sequence Data , Molecular Structure , Protein Binding , Purines/chemistry , Sequence Alignment , Sildenafil Citrate , Structure-Activity Relationship
16.
Eur J Med Chem ; 49: 200-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22309909

ABSTRACT

It is well known that 6-nitroquipazine exhibits about 150-fold higher affinity for the serotonin transporter (SERT) than quipazine and recently we showed quipazine buspirone analogues with high to moderate SERT affinity. Now we have designed and synthesized several 6-nitroquipazine buspirone derivatives. Unexpectedly, their SERT binding affinities were moderate, and much lower than that of the previously studied quipazine buspirone analogues. To explain these findings, docking studies of both groups of compounds into two different homology models of human SERT was performed using a flexible target-ligand docking approach (4D docking). The crystal structures of leucine transporter from Aquifex aeolicus in complex with leucine and with tryptophan were used as templates for the SERT models in closed and outward-facing conformations, respectively. We found that the latter conformation represents the most reliable model for binding of buspirone analogues. Docking into that model showed that the nitrated compounds acquire a rod like shape in the binding pocket with polar groups (nitro- and imido-) at the ends of the rod. 6-Nitro substituents gave steric clashes with amino acids located at the extracellular loop 4, which may explain their lower affinity than corresponding quipazine buspirone analogues. The results from the present study may suggest chemical design strategies to improve the SERT modulators.


Subject(s)
Buspirone/chemistry , Buspirone/pharmacology , Quipazine/analogs & derivatives , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Bacteria/chemistry , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Buspirone/chemical synthesis , Humans , Models, Molecular , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Quipazine/chemical synthesis , Quipazine/chemistry , Quipazine/pharmacology , Serotonin Plasma Membrane Transport Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/chemical synthesis
17.
Methods Mol Biol ; 857: 281-99, 2012.
Article in English | MEDLINE | ID: mdl-22323226

ABSTRACT

Transporter proteins are divided into channels and carriers and constitute families of membrane proteins of physiological and pharmacological importance. These proteins are targeted by several currently prescribed drugs, and they have a large potential as targets for new drug development. Ion channels and carriers are difficult to express and purify in amounts for X-ray crystallography and nuclear magnetic resonance (NMR) studies, and few carrier and ion channel structures are deposited in the PDB database. The scarcity of atomic resolution 3D structures of carriers and channels is a problem for understanding their molecular mechanisms of action and for designing new compounds with therapeutic potentials. The homology modeling approach is a valuable approach for obtaining structural information about carriers and ion channels when no crystal structure of the protein of interest is available. In this chapter, computational approaches for constructing homology models of carriers and transporters are reviewed.


Subject(s)
Carrier Proteins/chemistry , Ion Channels/metabolism , Structural Homology, Protein , ATP-Binding Cassette Transporters/chemistry , Animals , Humans , Models, Molecular , Sequence Alignment/methods
18.
J Mol Model ; 18(3): 1073-85, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21670993

ABSTRACT

The serotonin (5-HT) transporter (SERT) plays an important role in the termination of 5-HT-mediated neurotransmission by transporting 5-HT away from the synaptic cleft and into the presynaptic neuron. In addition, SERT is the main target for antidepressant drugs, including the selective serotonin reuptake inhibitors (SSRIs). The three-dimensional (3D) structure of SERT has not yet been determined, and little is known about the molecular mechanisms of substrate binding and transport, though such information is very important for the development of new antidepressant drugs. In this study, a homology model of SERT was constructed based on the 3D structure of a prokaryotic homologous leucine transporter (LeuT) (PDB id: 2A65). Eleven tryptamine derivates (including 5-HT) and the SSRI (S)-citalopram were docked into the putative substrate binding site, and two possible binding modes of the ligands were found. To study the conformational effect that ligand binding may have on SERT, two SERT-5-HT and two SERT-(S)-citalopram complexes, as well as the SERT apo structure, were embedded in POPC lipid bilayers and comparative molecular dynamics (MD) simulations were performed. Our results show that 5-HT in the SERT-5-HT(B) complex induced larger conformational changes in the cytoplasmic parts of the transmembrane helices of SERT than any of the other ligands. Based on these results, we suggest that the formation and breakage of ionic interactions with amino acids in transmembrane helices 6 and 8 and intracellular loop 1 may be of importance for substrate translocation.


Subject(s)
Molecular Dynamics Simulation , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin/chemistry , Computer Simulation , Humans , Protein Binding , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors , Substrate Specificity
19.
Eur J Med Chem ; 47(1): 24-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22071255

ABSTRACT

The two main groups of antidepressant drugs, the tricyclic antidepressants (TCAs) and the selective serotonin reuptake inhibitors (SSRIs), as well as several other compounds, act by inhibiting the serotonin transporter (SERT). However, the binding mode and molecular mechanism of inhibition in SERT are not fully understood. In this study, five classes of SERT inhibitors were docked into an outward-facing SERT homology model using a new 4D ensemble docking protocol. Unlike other docking protocols, where protein flexibility is not considered or is highly dependent on the ligand structure, flexibility was here obtained by side chain sampling of the amino acids of the binding pocket using biased probability Monte Carlo (BPMC) prior to docking. This resulted in the generation of multiple binding pocket conformations that the ligands were docked into. The docking results showed that the inhibitors were stacked between the aromatic amino acids of the extracellular gate (Y176, F335) presumably preventing its closure. The inhibitors interacted with amino acids in both the putative substrate binding site and more extracellular regions of the protein. A general structure-docking-based pharmacophore model was generated to explain binding of all studied classes of SERT inhibitors. Docking of a test set of actives and decoys furthermore showed that the outward-facing ensemble SERT homology model consistently and selectively scored the majority of active compounds above decoys, which indicates its usefulness in virtual screening.


Subject(s)
Models, Molecular , Selective Serotonin Reuptake Inhibitors/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/metabolism , Binding Sites , Drug Evaluation, Preclinical , Ligands , Mazindol/chemistry , Mazindol/metabolism , Mazindol/pharmacology , Monte Carlo Method , Protein Conformation , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins/chemistry , Selective Serotonin Reuptake Inhibitors/chemistry , Tropanes/chemistry , Tropanes/metabolism , Tropanes/pharmacology
20.
J Steroid Biochem Mol Biol ; 128(3-5): 154-64, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22051079

ABSTRACT

The aim of this study was to explore the effects of 22(S)-hydroxycholesterol (22(S)-HC) on lipid and glucose metabolism in human-derived cells from metabolic active tissues. Docking of T0901317 and 22(S)-HC showed that both substances fitted into the ligand binding domain of liver X receptors (LXR). Results show that while several lipogenic genes were induced by T0901317 in myotubes, HepG2 cells and SGBS cells, effect of 22(S)-HC varied more between cell types. In myotubes, most lipogenic genes were downregulated or unchanged by 22(S)-HC, whereas a more diverse pattern was found in HepG2 and SGBS cells. Treatment with 22(S)-HC induced sterol regulatory element binding transcription factor 1 in SGBS and HepG2 cells, but not in myotubes. Fatty acid synthase was downregulated by 22(S)-HC in myotubes, upregulated in SGBS and unchanged in HepG2 cells. De novo lipogenesis was increased by T0901317 in all cell models, whereas differently affected by 22(S)-HC depending on the cell type; decreased in myotubes and HepG2 cells, whereas increased in SGBS cells. Oxidation of linoleic acid was reduced by 22(S)-HC in all cell models while glucose uptake increased and tended to increase in myotubes and SGBS cells, respectively. Cholesterol efflux was unaffected by 22(S)-HC treatment. These results show that 22(S)-HC affects LXR-regulated processes differently in various cell types. Ability of 22(S)-HC to reduce lipogenesis and lipid accumulation in myotubes and hepatocytes indicate that 22(S)-HC might reduce lipid accumulation in non-adipose tissues, suggesting a potential role for 22(S)-HC or a similar LXR modulator in the treatment of type 2 diabetes.


Subject(s)
Anticholesteremic Agents/pharmacology , Glucose/metabolism , Hydroxycholesterols/pharmacology , Lipid Metabolism/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Anticholesteremic Agents/chemistry , Binding Sites , Cell Line , Cells, Cultured , Computational Biology/methods , Enterocytes/drug effects , Enterocytes/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydroxycholesterols/chemistry , Liver X Receptors , Models, Molecular , Molecular Conformation , Oligonucleotide Array Sequence Analysis , Organ Specificity , Orphan Nuclear Receptors/metabolism , RNA, Messenger/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...