Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
1.
Nature ; 597(7878): 698-702, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34526714

RÉSUMÉ

The development of new antibiotics to treat infections caused by drug-resistant Gram-negative pathogens is of paramount importance as antibiotic resistance continues to increase worldwide1. Here we describe a strategy for the rational design of diazabicyclooctane inhibitors of penicillin-binding proteins from Gram-negative bacteria to overcome multiple mechanisms of resistance, including ß-lactamase enzymes, stringent response and outer membrane permeation. Diazabicyclooctane inhibitors retain activity in the presence of ß-lactamases, the primary resistance mechanism associated with ß-lactam therapy in Gram-negative bacteria2,3. Although the target spectrum of an initial lead was successfully re-engineered to gain in vivo efficacy, its ability to permeate across bacterial outer membranes was insufficient for further development. Notably, the features that enhanced target potency were found to preclude compound uptake. An improved optimization strategy leveraged porin permeation properties concomitant with biochemical potency in the lead-optimization stage. This resulted in ETX0462, which has potent in vitro and in vivo activity against Pseudomonas aeruginosa plus all other Gram-negative ESKAPE pathogens, Stenotrophomonas maltophilia and biothreat pathogens. These attributes, along with a favourable preclinical safety profile, hold promise for the successful clinical development of the first novel Gram-negative chemotype to treat life-threatening antibiotic-resistant infections in more than 25 years.


Sujet(s)
Antibactériens/pharmacologie , Conception de médicament , Multirésistance bactérienne aux médicaments , Bactéries à Gram négatif/effets des médicaments et des substances chimiques , Animaux , Antibactériens/composition chimique , Composés aza/composition chimique , Composés aza/pharmacologie , Cyclooctanes/composition chimique , Cyclooctanes/pharmacologie , Femelle , Souris , Souris de lignée BALB C , Structure moléculaire , Protéines de liaison aux pénicillines/antagonistes et inhibiteurs , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , bêta-Lactamases
2.
Bioorg Med Chem ; 28(24): 115826, 2020 12 15.
Article de Anglais | MEDLINE | ID: mdl-33160146

RÉSUMÉ

UDP-3-O-(R-3-hydroxyacyl)-N-acetylglucosamine deacetylase (LpxC), the zinc metalloenzyme catalyzing the first committed step of lipid A biosynthesis in Gram-negative bacteria, has been a target for antibacterial drug discovery for many years. All inhibitor chemotypes reaching an advanced preclinical stage and clinical phase 1 have contained terminal hydroxamic acid, and none have been successfully advanced due, in part, to safety concerns, including hemodynamic effects. We hypothesized that the safety of LpxC inhibitors could be improved by replacing the terminal hydroxamic acid with a different zinc-binding group. After choosing an N-hydroxyformamide zinc-binding group, we investigated the structure-activity relationship of each part of the inhibitor scaffold with respect to Pseudomonas aeruginosa and Escherichia coli LpxC binding affinity, in vitro antibacterial potency and pharmacological properties. We identified a novel, potency-enhancing hydrophobic binding interaction for an LpxC inhibitor. We demonstrated in vivo efficacy of one compound in a neutropenic mouse E. coli infection model. Another compound was tested in a rat hemodynamic assay and was found to have a hypotensive effect. This result demonstrated that replacing the terminal hydroxamic acid with a different zinc-binding group was insufficient to avoid this previously recognized safety issue with LpxC inhibitors.


Sujet(s)
Amidohydrolases/antagonistes et inhibiteurs , Antibactériens/pharmacologie , Antienzymes/composition chimique , Formamides/composition chimique , Hémodynamique/effets des médicaments et des substances chimiques , Amidohydrolases/métabolisme , Animaux , Antibactériens/composition chimique , Antibactériens/métabolisme , Antibactériens/usage thérapeutique , Sites de fixation , Cristallographie aux rayons X , Modèles animaux de maladie humaine , Antienzymes/métabolisme , Antienzymes/pharmacologie , Antienzymes/usage thérapeutique , Escherichia coli/effets des médicaments et des substances chimiques , Infections à Escherichia coli/traitement médicamenteux , Infections à Escherichia coli/anatomopathologie , Femelle , Formamides/métabolisme , Formamides/pharmacologie , Formamides/usage thérapeutique , Période , Mâle , Souris , Simulation de dynamique moléculaire , Rats , Rat Sprague-Dawley , Relation structure-activité
3.
ACS Infect Dis ; 5(6): 863-872, 2019 06 14.
Article de Anglais | MEDLINE | ID: mdl-30848883

RÉSUMÉ

The high-molecular mass penicillin-binding proteins (PBPs) are the essential targets of the ß-lactam classes of antibacterial drugs. In the Gram-negative pathogen Escherichia coli, these include PBP1a, PBP1b, PBP2, and PBP3. Techniques that enable facile measurement of the potency of inhibition of these targets are valuable for understanding structure-activity relationships in programs aimed at discovering new antibiotics to combat drug-resistant infections. Continuous fluorescence anisotropy-based assays for inhibition of soluble constructs of PBP1a, PBP2, and PBP3 from the serious Gram-negative bacterial pathogens Pseudomonas aeruginosa and Acinetobacter baumannii and PBP3 from E. coli using the fluorescent phenoxypenicillin analogue BOCILLIN FL have been described previously, but this technique was not useful for PBP2 from E. coli due to a lack of change in fluorescence anisotropy or intensity upon reaction. Here, we report that a fluorescent analogue of ampicillin, 5-carboxytetramethylrhodamine-ampicillin (5-TAMRA-ampicillin), was useful as the indicator in a continuous fluorescence anisotropy-based kinetic assay for inhibition of a soluble construct of PBP2 from E. coli. The assay enables measurement of the bimolecular rate constant for inhibition kinact /Ki. This measurement was made for representative drugs from four classes of ß-lactams and for the diazabicyclooctenone ETX2514. 5-TAMRA-ampicillin was also useful in a fluorescence anisotropy-based assay for P. aeruginosa PBP2 and in fluorescence intensity-based assays with PBP1a and PBP3 from P. aeruginosa and A. baumannii and PBP3 from E. coli.


Sujet(s)
Ampicilline/pharmacologie , Protéines Escherichia coli/antagonistes et inhibiteurs , Escherichia coli/effets des médicaments et des substances chimiques , Protéines de liaison aux pénicillines/antagonistes et inhibiteurs , Peptidyl transferases/antagonistes et inhibiteurs , Rhodamines/pharmacologie , Acinetobacter baumannii/enzymologie , Ampicilline/analogues et dérivés , Antibactériens/pharmacologie , Escherichia coli/enzymologie , Polarisation de fluorescence , Cinétique , Tests de sensibilité microbienne , Pseudomonas aeruginosa/enzymologie , bêta-Lactames/pharmacologie
4.
Beilstein J Org Chem ; 14: 1750-1757, 2018.
Article de Anglais | MEDLINE | ID: mdl-30112080

RÉSUMÉ

The 1,3-dithiane-based dM-Dmoc group was studied for the protection of amino groups. Protection was achieved under mild conditions for aliphatic amines, and under highly reactive conditions for the less reactive arylamines. Moderate to excellent yields were obtained. Deprotection was performed by oxidation followed by treating with a weak base. The yields were good to excellent. The new amino protecting group offers a different dimension of orthogonality in reference to the commonly used amino protecting groups in terms of deprotection conditions. It is expected to allow a collection of transformations to be carried out on the protected substrates that are unattainable using any known protecting groups.

5.
ACS Infect Dis ; 3(4): 310-319, 2017 04 14.
Article de Anglais | MEDLINE | ID: mdl-28157293

RÉSUMÉ

The global emergence of antibiotic resistance, especially in Gram-negative bacteria, is an urgent threat to public health. Discovery of novel classes of antibiotics with activity against these pathogens has been impeded by a fundamental lack of understanding of the molecular drivers underlying small molecule uptake. Although it is well-known that outer membrane porins represent the main route of entry for small, hydrophilic molecules across the Gram-negative cell envelope, the structure-permeation relationship for porin passage has yet to be defined. To address this knowledge gap, we developed a sensitive and specific whole-cell approach in Escherichia coli called titrable outer membrane permeability assay system (TOMAS). We used TOMAS to characterize the structure porin-permeation relationships of a set of novel carbapenem analogues through the Pseudomonas aeruginosa porin OprD. Our results show that small structural modifications, especially the number and nature of charges and their position, have dramatic effects on the ability of these molecules to permeate cells through OprD. This is the first demonstration of a defined relationship between specific molecular changes in a substrate and permeation through an isolated porin. Understanding the molecular mechanisms that impact antibiotic transit through porins should provide valuable insights to antibacterial medicinal chemistry and may ultimately allow for the rational design of porin-mediated uptake of small molecules into Gram-negative bacteria.


Sujet(s)
Carbapénèmes/composition chimique , Porines/métabolisme , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , Carbapénèmes/pharmacologie , Résistance microbienne aux médicaments/effets des médicaments et des substances chimiques , Escherichia coli/génétique , Escherichia coli/croissance et développement , Tests de sensibilité microbienne , Porines/génétique , Pseudomonas aeruginosa/métabolisme , Relation structure-activité
6.
Bioorg Med Chem Lett ; 26(19): 4775-4780, 2016 10 01.
Article de Anglais | MEDLINE | ID: mdl-27578247

RÉSUMÉ

During the lead generation and optimization of PARP inhibitors blocking centrosome clustering, it was discovered that increasing hydrogen bond acceptor (HBA) strength improved cellular potency but led to elevated Caco2 and MDR1 efflux and thus poor oral bioavailability. Conversely, compounds with lower efflux had reduced potency. The project team was able to improve the bioavailability by reducing efflux through systematic modifications to the strength of the HBA by changing the electronic properties of neighboring groups, whilst maintaining sufficient acceptor strength for potency. Additionally, it was observed that enantiomers with different potency showed similar efflux, which is consistent with the promiscuity of efflux transporters. Eventually, a balance between potency and low efflux was achieved for a set of lead compounds with good bioavailability which allowed the project to progress towards establishing in vivo pharmacokinetic/pharmacodynamic relationships.


Sujet(s)
Centrosome/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacocinétique , Administration par voie orale , Animaux , Biodisponibilité , Cellules Caco-2 , Chiens , Humains , Liaison hydrogène , Cellules rénales canines Madin-Darby , Souris , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Rats
7.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-26546219

RÉSUMÉ

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Sujet(s)
Centrosome/métabolisme , Phtalazines/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Administration par voie orale , Animaux , Sites de fixation , Cellules Caco-2 , Centrosome/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Évaluation préclinique de médicament , Cellules HeLa , Humains , Microsomes/métabolisme , Conformation moléculaire , Simulation de dynamique moléculaire , Phtalazines/administration et posologie , Phtalazines/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/administration et posologie , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Structure tertiaire des protéines , Rats , Tankyrases/antagonistes et inhibiteurs , Tankyrases/métabolisme
8.
ACS Med Chem Lett ; 6(5): 537-42, 2015 May 14.
Article de Anglais | MEDLINE | ID: mdl-26005529

RÉSUMÉ

A main challenge in the development of new agents for the treatment of Pseudomonas aeruginosa infections is the identification of chemotypes that efficiently penetrate the cell envelope and are not susceptible to established resistance mechanisms. Siderophore-conjugated monocarbams are attractive because of their ability to hijack the bacteria's iron uptake machinery for transport into the periplasm and their inherent stability to metallo-ß-lactamases. Through development of the SAR we identified a number of modifications to the scaffold that afforded active anti-P. aeruginosa agents with good physicochemical properties. Through crystallographic efforts we gained a better understanding into how these compounds bind to the target penicillin binding protein PBP3 and factors to consider for future design.

9.
Chem Biol ; 22(4): 535-547, 2015 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-25910245

RÉSUMÉ

Understanding how compound penetration occurs across the complex cell walls of Gram-negative bacteria is one of the greatest challenges in discovering new drugs to treat the infections they cause. A combination of next-generation transposon sequencing, computational metadynamics simulations (CMDS), and medicinal chemistry was used to define genetic and structural elements involved in facilitated carbapenem entry into Pseudomonas aeruginosa. Here we show for the first time that these compounds are taken up not only by the major outer membrane channel OccD1 (also called OprD or PA0958) but also by a closely related channel OccD3 (OpdP or PA4501). Transport-mediating molecular interactions predicted by CMDS for these channels were first confirmed genetically, then used to guide the design of carbapenem analogs with altered uptake properties. These results bring us closer to the rational design of channel transmissibility and may ultimately lead to improved permeability of compounds across bacterial outer membranes.


Sujet(s)
Carbapénèmes/métabolisme , Pseudomonas aeruginosa/métabolisme , Substitution d'acide aminé , Protéines de la membrane externe bactérienne/composition chimique , Protéines de la membrane externe bactérienne/génétique , Protéines de la membrane externe bactérienne/métabolisme , Sites de fixation , Carbapénèmes/composition chimique , Carbapénèmes/pharmacologie , Tests de sensibilité microbienne , Simulation de docking moléculaire , Structure tertiaire des protéines , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , Spécificité du substrat
10.
J Med Chem ; 58(5): 2195-205, 2015 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-25658376

RÉSUMÉ

To identify new agents for the treatment of multi-drug-resistant Pseudomonas aeruginosa, we focused on siderophore-conjugated monocarbams. This class of monocyclic ß-lactams are stable to metallo-ß-lactamases and have excellent P. aeruginosa activities due to their ability to exploit the iron uptake machinery of Gram-negative bacteria. Our medicinal chemistry plan focused on identifying a molecule with optimal potency and physical properties and activity for in vivo efficacy. Modifications to the monocarbam linker, siderophore, and oxime portion of the molecules were examined. Through these efforts, a series of pyrrolidinone-based monocarbams with good P. aeruginosa cellular activity (P. aeruginosa MIC90 = 2 µg/mL), free fraction levels (>20% free), and hydrolytic stability (t1/2 ≥ 100 h) were identified. To differentiate the lead compounds and enable prioritization for in vivo studies, we applied a semi-mechanistic pharmacokinetic/pharmacodynamic model to enable prediction of in vivo efficacy from in vitro data.


Sujet(s)
Antibactériens/pharmacologie , Antibactériens/pharmacocinétique , Découverte de médicament , Monobactames/pharmacologie , Monobactames/pharmacocinétique , Infections à Pseudomonas/traitement médicamenteux , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , Sidérophores/métabolisme , Animaux , Humains , Mâle , Monobactames/composition chimique , Infections à Pseudomonas/microbiologie , Rats , Rat Wistar , Relation structure-activité , bêta-Lactamases/composition chimique
11.
J Biomol Screen ; 19(8): 1137-46, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-24820111

RÉSUMÉ

A novel, ultrahigh-throughput, fluorescence anisotropy-based assay was developed and used to screen a 1.4-million-sample library for compounds that compete with adenosine triphosphate (ATP) for binding to Escherichia coli tRNA(Ile) lysidine synthetase (TilS), an essential, conserved, ATP-dependent, tRNA-modifying enzyme of bacterial pathogens. TilS modifies a cytidine base in the anticodon loop of Ile2 tRNA by attaching lysine, thereby altering codon recognition of the CAU anticodon from AUG (methionine) to AUA (isoleucine). A scintillation proximity assay for the incorporation of lysine into Ile2 tRNA was used to eliminate false positives in the initial screen resulting from detection artifacts as well as compounds competitive with the fluorescent label instead of ATP, and to measure inhibitor potencies against E. coli and Pseudomonas aeruginosa TilS isozymes. The tRNA(Ile) substrate for P. aeruginosa TilS was identified for the first time to enable these measurements. ATP-competitive binding of inhibitors was confirmed by one-dimensional ligand-observe nuclear magnetic resonance. A preliminary structure-activity relationship is shown for two inhibitor series.


Sujet(s)
Amino acyl-tRNA synthetases/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Protéines Escherichia coli/antagonistes et inhibiteurs , Tests de criblage à haut débit/méthodes , Adénosine triphosphate/métabolisme , Amino acyl-tRNA synthetases/génétique , Amino acyl-tRNA synthetases/métabolisme , Anticodon , Fixation compétitive , Antienzymes/composition chimique , Antienzymes/métabolisme , Protéines Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Polarisation de fluorescence/méthodes , Lysine/métabolisme , Pseudomonas aeruginosa/enzymologie , Relation structure-activité
12.
J Med Chem ; 57(3): 733-58, 2014 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-24410637

RÉSUMÉ

A new series of potent and selective histamine-3 receptor (H3R) antagonists was identified on the basis of an azaspiro[2.5]octane carboxamide scaffold. Many scaffold modifications were largely tolerated, resulting in nanomolar-potent compounds in the H3R functional assay. Exemplar compound 6s demonstrated a selective profile against a panel of 144 secondary pharmacological receptors, with activity at only σ2 (62% at 10 µM). Compound 6s demonstrated free-plasma exposures above the IC50 (∼50×) with a brain-to-plasma ratio of ∼3 following intravenous dosing in mice. At three doses tested in the mouse novel object recognition model (1, 3, and 10 mg/kg s.c.), 6s demonstrated a statistically significant response compared with the control group. This series represents a new scaffold of H3 receptor antagonists that demonstrates in vivo exposure and efficacy in an animal model of cognition.


Sujet(s)
Cognition/effets des médicaments et des substances chimiques , Cyclopropanes/synthèse chimique , Antihistaminiques des récepteurs H3/synthèse chimique , Pipérazines/synthèse chimique , Récepteur histaminergique H3/métabolisme , Spiranes/synthèse chimique , Animaux , Azétidines/synthèse chimique , Azétidines/pharmacocinétique , Azétidines/pharmacologie , Cellules CHO , Perméabilité des membranes cellulaires , Cricetinae , Cricetulus , Cyclopropanes/pharmacocinétique , Cyclopropanes/pharmacologie , Chiens , Antihistaminiques des récepteurs H3/pharmacocinétique , Antihistaminiques des récepteurs H3/pharmacologie , Humains , Apprentissage/effets des médicaments et des substances chimiques , Cellules rénales canines Madin-Darby , Mâle , Souris , Microsomes du foie/métabolisme , Modèles moléculaires , Pipérazines/pharmacocinétique , Pipérazines/pharmacologie , Pipéridines/synthèse chimique , Pipéridines/pharmacocinétique , Pipéridines/pharmacologie , Pyrrolidines/synthèse chimique , Pyrrolidines/pharmacocinétique , Pyrrolidines/pharmacologie , Rats , Rat Sprague-Dawley , Récepteur histaminergique H3/génétique , /effets des médicaments et des substances chimiques , Spiranes/pharmacocinétique , Spiranes/pharmacologie , Stéréoisomérie , Relation structure-activité
13.
Bioorg Med Chem Lett ; 21(11): 3399-403, 2011 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-21524576

RÉSUMÉ

Herein we describe the discovery of compounds that are competitive antagonists of the CP101-606 binding site within the NR2B subtype of the NMDA receptor. The compounds identified do not possess phenolic functional groups such as those in ifenprodil and related analogs. Initial identification of hits in this series focused on a basic, secondary amine side chain which led to good potency, but also presented a hERG liability. Further modifications led to examples of non-basic replacements which demonstrated much less liability in this regard. Finally, one compound in the series, 6a, was tested in the mouse forced swim depression assay and found to show activity (s.c. 60 mg/kg).


Sujet(s)
Antidépresseurs/synthèse chimique , Pyrazines/synthèse chimique , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Animaux , Antidépresseurs/composition chimique , Antidépresseurs/pharmacologie , Sites de fixation , Fixation compétitive , Relation dose-effet des médicaments , Concentration inhibitrice 50 , Souris , Structure moléculaire , Activité motrice/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques , Pyrazines/composition chimique , Pyrazines/pharmacologie
14.
Curr Pharm Des ; 17(12): 1188-92, 2011.
Article de Anglais | MEDLINE | ID: mdl-21492088

RÉSUMÉ

Obesity and substance use disorders are rapidly growing problems throughout the world. Of the current mainstay therapies of diet, exercise, behavioral modification, surgery, and medications, drugs have the greatest risk for abuse and dependence. As each of these disorders share similar underpinnings mediated by the dopaminergic brain reward pathways, clinicians must seriously consider the safety of both the patient's physical and mental health when prescribing treatments. Specifically, balance and awareness of the factors involved in the variable abuse potentials of these prescribed medications is paramount. A cursory review of weight loss medications commonly used is performed with attention to FDA status, mechanism of action, and abuse potential. Concurrent strategies to minimize risk such as drug screening, ruling out doctor shopping, temporal considerations, monitoring for signs and symptoms of abuse and/or dependency, and a safety-tiered prescribing approach is also discussed in order to optimize best treatment practice. As the understanding of these disorders progresses along with the evolution of agreed nomenclature and awareness of compulsive behavioral disorders in general, greater safety and more appropriate interventions may be achieved. Further areas of research will help to elucidate nuances of the coocurrance and treatment of these disorders and perhaps guide drug research and development in the area of drug treatments of obesity.


Sujet(s)
Neuroleptiques/usage thérapeutique , Thérapie comportementale , Comportement toxicomaniaque/traitement médicamenteux , Obésité/traitement médicamenteux , Animaux , Humains , Facteurs de risque
15.
Eur J Pharmacol ; 649(1-3): 59-63, 2010 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-20854803

RÉSUMÉ

The peptide corticotropin-releasing factor (CRF) binds to the CRF1 receptor via a two-domain mechanism such that the extracellular domain (ECD) of the receptor captures the CRF's C-terminus to facilitate the binding of CRF's N-terminus to the juxta-membrane or "J"-site. Known small molecule antagonists bind to the J-site while known CRF1 receptor peptide radioligands bind to both sites. We report here the in vitro binding properties of the first radioligand that binds exclusively to the ECD of the CRF1 receptor. This ligand, which we named [¹²5I]Yamada peptide 20 ([¹²5I]YP20), is a radiolabeled analog of a synthetic peptide first reported by Yamada et al. (2004). We confirmed its high affinity for the [¹²5I]CRF binding site on the hCRF1 receptor and also found it to potently antagonize CRF-stimulated cAMP production in hCRF1-CHO cells. Under optimized conditions, 20 pM [¹²5I]YP20 reproducibly bound to hCRF1-CHO membranes with a pharmacology consistent with binding specific to the ECD of the CRF1 receptor. Saturation binding studies revealed the presence of a high affinity site with an estimated K(d) of ≈0.9 nM. The kinetic association of 20 pM [¹²5I]YP20 binding best fit to a rapid component (t(1/2)=0.69 min) and a sluggish component (t(1/2)=42 min). [¹²5I]YP20's specific binding was rapidly reversible with dissociation kinetics also best described by two phases (t(1/2)=0.92 min and t(1/2)=11.7 min). While [¹²5I]YP20's binding kinetics are complex, its high affinity and pharmacological specificity indicate that it is an excellent radioligand for probing the ECD site of the CRF1 receptor.


Sujet(s)
Corticolibérine/antagonistes et inhibiteurs , Oligopeptides/métabolisme , Oligopeptides/pharmacologie , Peptides/métabolisme , Peptides/pharmacologie , Motifs et domaines d'intéraction protéique , Récepteur CRH/métabolisme , Adenylate Cyclase/métabolisme , Animaux , Sites de fixation , Fixation compétitive/effets des médicaments et des substances chimiques , Cellules CHO , Corticolibérine/métabolisme , Cricetinae , Cricetulus , AMP cyclique/métabolisme , Évaluation préclinique de médicament/méthodes , Activation enzymatique/effets des médicaments et des substances chimiques , Guanylyl imidodiphosphate/métabolisme , Humains , Radio-isotopes de l'iode , Cinétique , Ligands , Peptides/antagonistes et inhibiteurs , Pyrimidines/métabolisme , Récepteur CRH/antagonistes et inhibiteurs , Récepteur CRH/biosynthèse , Récepteur CRH/génétique , Protéines recombinantes/biosynthèse , Protéines recombinantes/métabolisme
16.
J Med Chem ; 50(24): 5912-25, 2007 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-17985862

RÉSUMÉ

Fragment-based lead generation has led to the discovery of a novel series of cyclic amidine-based inhibitors of beta-secretase (BACE-1). Initial fragment hits with an isocytosine core having millimolar potency were identified via NMR affinity screening. Structure-guided evolution of these fragments using X-ray crystallography together with potency determination using surface plasmon resonance and functional enzyme inhibition assays afforded micromolar inhibitors. Similarity searching around the isocytosine core led to the identification of a related series of inhibitors, the dihydroisocytosines. By leveraging the knowledge of the ligand-BACE-1 recognition features generated from the isocytosines, the dihydroisocytosines were efficiently optimized to submicromolar potency. Compound 29, with an IC50 of 80 nM, a ligand efficiency of 0.37, and cellular activity of 470 nM, emerged as the lead structure for future optimization.


Sujet(s)
Amidines/synthèse chimique , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Aspartic acid endopeptidases/antagonistes et inhibiteurs , Cytosine/analogues et dérivés , Modèles moléculaires , Pyrimidines/synthèse chimique , Amidines/composition chimique , Amidines/pharmacologie , Amyloid precursor protein secretases/composition chimique , Amyloid precursor protein secretases/génétique , Aspartic acid endopeptidases/composition chimique , Aspartic acid endopeptidases/génétique , Lignée cellulaire , Cristallographie aux rayons X , Cytosine/synthèse chimique , Cytosine/composition chimique , Cytosine/pharmacologie , Transfert d'énergie par résonance de fluorescence , Humains , Ligands , Spectroscopie par résonance magnétique , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Stéréoisomérie , Relation structure-activité
17.
J Biomech ; 36(11): 1659-68, 2003 Nov.
Article de Anglais | MEDLINE | ID: mdl-14522207

RÉSUMÉ

Computational wear prediction is an attractive concept for evaluating new total knee replacement designs prior to physical testing and implementation. An important hurdle to such technology is the lack of in vivo contact pressure predictions. To address this issue, this study evaluates a computationally efficient simulation approach that combines the advantages of rigid and deformable body modeling. The hybrid method uses rigid body dynamics to predict body positions and orientations and elastic foundation theory to predict contact pressures between general three-dimensional surfaces. To evaluate the method, we performed static pressure experiments with a commercial knee implant in neutral alignment using flexion angles of 0, 30, 60, and 90 degrees and loads of 750, 1500, 2250, and 3000N. Using manufacturer CAD geometry for the same implant, an elastic foundation model with linear or nonlinear polyethylene material properties was implemented within a commercial multibody dynamics software program. The model's ability to predict experimental peak and average contact pressures simultaneously was evaluated by performing dynamic simulations to find the static configuration. Both the linear and nonlinear material models predicted the average contact pressure data well, while only the linear material model could simultaneously predict the trends in the peak contact pressure data. This novel modeling approach is sufficiently fast and accurate to be used in design sensitivity and optimization studies of knee implant mechanics and ultimately wear.


Sujet(s)
Analyse de panne d'appareillage/méthodes , Articulation du genou/physiopathologie , Articulation du genou/chirurgie , Prothèse de genou , Test de matériaux/méthodes , Modèles biologiques , Mise en charge , Résistance à la compression , Élasticité , Humains , Dynamique non linéaire , Pression , Reproductibilité des résultats , Sensibilité et spécificité , Propriétés de surface
18.
J Biol Chem ; 277(35): 31499-505, 2002 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-12072428

RÉSUMÉ

Cerebral deposition of amyloid beta-protein (A beta) is believed to play a key role in the pathogenesis of Alzheimer's disease. Because A beta is produced from the processing of amyloid beta-protein precursor (APP) by beta- and gamma-secretases, these enzymes are considered important therapeutic targets for identification of drugs to treat Alzheimer's disease. Unlike beta-secretase, which is a monomeric aspartyl protease, gamma-secretase activity resides as part of a membrane-bound, high molecular weight, macromolecular complex. Pepstatin and L685458 are among several structural classes of gamma-secretase inhibitors identified so far. These compounds possess a hydroxyethylene dipeptide isostere of aspartyl protease transition state analogs, suggesting gamma-secretase may be an aspartyl protease. However, the mechanism of inhibition of gamma-secretase by pepstatin and L685458 has not been elucidated. In this study, we report that pepstatin A methylester and L685458 unexpectedly displayed linear non-competitive inhibition of gamma-secretase. Sulfonamides and benzodiazepines, which do not resemble transition state analogs of aspartyl proteases, also displayed potent, non-competitive inhibition of gamma-secretase. Models to rationalize how transition state analogs inhibit their targets by non-competitive inhibition are discussed.


Sujet(s)
Benzodiazépines/pharmacologie , Carbamates/pharmacologie , Dipeptides/pharmacologie , Endopeptidases/métabolisme , Pepstatines/pharmacologie , Inhibiteurs de protéases/pharmacologie , Sulfonamides/pharmacologie , Amyloid precursor protein secretases , Aspartic acid endopeptidases , Sites de fixation , Humains , Cinétique , Modèles moléculaires , Protéines recombinantes/antagonistes et inhibiteurs
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE