Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Chem Rev ; 117(12): 8094-8128, 2017 Jun 28.
Article in English | MEDLINE | ID: mdl-28541045

ABSTRACT

Peptides and proteins are not orally bioavailable in mammals, although a few peptides are intestinally absorbed in small amounts. Polypeptides are generally too large and polar to passively diffuse through lipid membranes, while most known active transport mechanisms facilitate cell uptake of only very small peptides. Systematic evaluations of peptides with molecular weights above 500 Da are needed to identify parameters that influence oral bioavailability. Here we describe 125 cyclic peptides containing four to thirty-seven amino acids that are orally absorbed by mammals. Cyclization minimizes degradation in the gut, blood, and tissues by removing cleavable N- and C-termini and by shielding components from metabolic enzymes. Cyclization also folds peptides into bioactive conformations that determine exposure of polar atoms to solvation by water and lipids and therefore can influence oral bioavailability. Key chemical properties thought to influence oral absorption and bioavailability are analyzed, including molecular weight, octanol-water partitioning, hydrogen bond donors/acceptors, rotatable bonds, and polar surface area. The cyclic peptides violated to different degrees all of the limits traditionally considered to be important for oral bioavailability of drug-like small molecules, although fewer hydrogen bond donors and reduced flexibility generally favored oral absorption.


Subject(s)
Absorption, Physicochemical , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Humans , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism
2.
Sci Rep ; 6: 24575, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-27094554

ABSTRACT

Drug discovery and translation are normally based on optimizing efficacy by increasing receptor affinity, functional potency, drug-likeness (rule-of-five compliance) and oral bioavailability. Here we demonstrate that residence time of a compound on its receptor has an overriding influence on efficacy, exemplified for antagonists of inflammatory protein complement C5a that activates immune cells and promotes disease. Three equipotent antagonists (3D53, W54011, JJ47) of inflammatory responses to C5a (3 nM) were compared for drug-likeness, receptor affinity and antagonist potency in human macrophages, and anti-inflammatory efficacy in rats. Only the least drug-like antagonist (3D53) maintained potency in cells against higher C5a concentrations and had a much longer duration of action (t1/2 ~ 20 h) than W54011 or JJ47 (t1/2 ~ 1 -3 h) in inhibiting macrophage responses. The unusually long residence time of 3D53 on its receptor was mechanistically probed by molecular dynamics simulations, which revealed long-lasting interactions that trap the antagonist within the receptor. Despite negligible oral bioavailability, 3D53 was much more orally efficacious than W54011 or JJ47 in preventing repeated agonist insults to induce rat paw oedema over 24 h. Thus, residence time on a receptor can trump drug-likeness in determining efficacy, even oral efficacy, of pharmacological agents.


Subject(s)
Complement C5a/antagonists & inhibitors , Complement C5a/metabolism , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Receptor, Anaphylatoxin C5a/metabolism , Animals , Biological Availability , Chemotaxis/drug effects , Chemotaxis/immunology , Complement C5a/immunology , Disease Models, Animal , Edema/drug therapy , Edema/immunology , Edema/metabolism , Humans , Immunosuppressive Agents/chemistry , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Rats , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/chemistry
3.
PLoS One ; 10(7): e0133805, 2015.
Article in English | MEDLINE | ID: mdl-26225423

ABSTRACT

Antibacterial drugs with novel scaffolds and new mechanisms of action are desperately needed to address the growing problem of antibiotic resistance. The periplasmic oxidative folding system in Gram-negative bacteria represents a possible target for anti-virulence antibacterials. By targeting virulence rather than viability, development of resistance and side effects (through killing host native microbiota) might be minimized. Here, we undertook the design of peptidomimetic inhibitors targeting the interaction between the two key enzymes of oxidative folding, DsbA and DsbB, with the ultimate goal of preventing virulence factor assembly. Structures of DsbB--or peptides--complexed with DsbA revealed key interactions with the DsbA active site cysteine, and with a hydrophobic groove adjacent to the active site. The present work aimed to discover peptidomimetics that target the hydrophobic groove to generate non-covalent DsbA inhibitors. The previously reported structure of a Proteus mirabilis DsbA active site cysteine mutant, in a non-covalent complex with the heptapeptide PWATCDS, was used as an in silico template for virtual screening of a peptidomimetic fragment library. The highest scoring fragment compound and nine derivatives were synthesized and evaluated for DsbA binding and inhibition. These experiments discovered peptidomimetic fragments with inhibitory activity at millimolar concentrations. Although only weakly potent relative to larger covalent peptide inhibitors that interact through the active site cysteine, these fragments offer new opportunities as templates to build non-covalent inhibitors. The results suggest that non-covalent peptidomimetics may need to interact with sites beyond the hydrophobic groove in order to produce potent DsbA inhibitors.


Subject(s)
Bacterial Proteins/metabolism , Oxidoreductases/metabolism , Peptides/pharmacology , Peptidomimetics/pharmacology , Protein Disulfide-Isomerases/metabolism , Toluene/analogs & derivatives , Catalytic Domain/drug effects , Cysteine/metabolism , Proteus mirabilis/drug effects , Proteus mirabilis/metabolism , Toluene/metabolism , Virulence/drug effects , Virulence Factors/metabolism
4.
J Chem Inf Model ; 55(6): 1181-91, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26000704

ABSTRACT

Protease activated receptor 2 (PAR2) is an unusual G-protein coupled receptor (GPCR) involved in inflammation and metabolism. It is activated through cleavage of its N-terminus by proteases. The new N-terminus functions as a tethered ligand that folds back and intramolecularly activates PAR2, initiating multiple downstream signaling pathways. The only compounds reported to date to inhibit PAR2 activation are of moderate potency. Three structural models for PAR2 have been constructed based on sequence homology with known crystal structures for bovine rhodopsin, human ORL-1 (also called nociceptin/orphanin FQ receptor), and human PAR1. The three PAR2 model structures were compared and used to predict potential interactions with ligands. Virtual screening for ligands using the Chembridge database, and either ORL-1 or PAR1 derived PAR2 models led to identification of eight new small molecule PAR2 antagonists (IC50 10-100 µM). Notably, the most potent compound 1 (IC50 11 µM) was derived from the less homologous template protein, human ORL-1. The results suggest that virtual screening against multiple homology models of the same GPCR can produce structurally diverse antagonists and that this may be desirable even when some models have less sequence homology with the target protein.


Subject(s)
Drug Discovery/methods , Molecular Docking Simulation , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/chemistry , Sequence Homology, Amino Acid , Animals , Binding Sites , Cattle , Cell Membrane/metabolism , Databases, Protein , Drug Evaluation, Preclinical , HT29 Cells , Humans , Ligands , Protein Structure, Tertiary , Receptor, PAR-2/metabolism
5.
ACS Med Chem Lett ; 5(10): 1148-51, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25313329

ABSTRACT

Development of peptide-based drugs has been severely limited by lack of oral bioavailability with less than a handful of peptides being truly orally bioavailable, mainly cyclic peptides with N-methyl amino acids and few hydrogen bond donors. Here we report that cyclic penta- and hexa-leucine peptides, with no N-methylation and five or six amide NH protons, exhibit some degree of oral bioavailability (4-17%) approaching that of the heavily N-methylated drug cyclosporine (22%) under the same conditions. These simple cyclic peptides demonstrate that oral bioavailability is achievable for peptides that fall outside of rule-of-five guidelines without the need for N-methylation or modified amino acids.

6.
J Med Chem ; 57(20): 8459-70, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25259874

ABSTRACT

The G-protein coupled receptor (C3aR) for human inflammatory protein complement C3a is an important component of immune, inflammatory, and metabolic diseases. A flexible compound (N2-[(2,2-diphenylethoxy)acetyl]-l-arginine, 4), known as a weak C3aR antagonist (IC50 µM), was transformed here into potent agonists (EC50 nM) of human macrophages (Ca(2+) release in HMDM) by incorporating aromatic heterocycles. Antagonists were also identified. A linear correlation between binding affinity for C3aR and calculated hydrogen-bond interaction energy of the heteroatom indicated that its hydrogen-bonding capacity influenced ligand affinity and function mediated by C3aR. Hydrogen-bond accepting heterocycles (e.g., imidazole) conferred the highest affinity and agonist potency (e.g., 21, EC50 24 nM, Ca(2+), HMDM) with comparable efficacy and immunostimulatory activity as that of C3a in activating human macrophages (Ca(2+), IL1ß, TNFα, CCL3). These potent and selective modulators of C3aR, inactivated by a C3aR antagonist, are stable C3a surrogates for interrogating roles for C3aR in physiology and disease.


Subject(s)
Heterocyclic Compounds/chemistry , Receptors, Complement/agonists , Receptors, Complement/metabolism , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Arginine/analogs & derivatives , Arginine/chemistry , Arginine/pharmacology , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Calcium/metabolism , Cells, Cultured , Chemistry Techniques, Synthetic , Gene Expression Regulation/drug effects , Heterocyclic Compounds/pharmacology , Humans , Hydrogen Bonding , Ligands , Macrophages/drug effects , Macrophages/metabolism , Receptors, Complement/antagonists & inhibitors , Structure-Activity Relationship
7.
Nat Commun ; 4: 2802, 2013.
Article in English | MEDLINE | ID: mdl-24257095

ABSTRACT

A significant challenge in chemistry is to rationally reproduce the functional potency of a protein in a small molecule, which is cheaper to manufacture, non-immunogenic, and also both stable and bioavailable. Synthetic peptides corresponding to small bioactive protein surfaces do not form stable structures in water and do not exhibit the functional potencies of proteins. Here we describe a novel approach to growing small molecules with protein-like potencies from a functionally important amino acid of a protein. A 77-residue human inflammatory protein (complement C3a) important in innate immunity is rationally transformed to equipotent small molecules, using peptide surrogates that incorporate a turn-inducing heterocycle with correctly positioned hydrogen-bond-accepting atoms. Small molecule agonists (molecular weight <500 Da) examined for receptor affinity and cellular responses have the same high potencies, functional profile and specificity of action as C3a protein, but greater plasma stability and bioavailability.


Subject(s)
Complement C3a/chemistry , Complement C3a/physiology , Inflammation Mediators/chemistry , Inflammation Mediators/physiology , Receptors, Complement/agonists , Complement C3a/metabolism , Humans , Inflammation Mediators/metabolism , Structure-Activity Relationship
8.
J Gen Virol ; 94(Pt 9): 1961-1971, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23740481

ABSTRACT

The flavivirus nonstructural protein 5 (NS5) is a large protein that is structurally conserved among members of the genus, making it an attractive target for antiviral drug development. The protein contains a methyltransferase (MTase) domain and an RNA dependent RNA polymerase (POL) domain. Previous studies with dengue viruses have identified a genetic interaction between residues 46-49 in the αA3-motif in the MTase and residue 512 in POL. These genetic interactions are consistent with structural modelling of these domains in West Nile virus (WNV) NS5 that predict close proximity of these regions of the two domains, and potentially a functional interaction mediated via the αA3-motif. To demonstrate an interaction between the MTase and POL domains of the WNV NS5 protein, we co-expressed affinity-tagged recombinant MTase and POL proteins in human embryonic kidney cells with simian virus 40 large T antigen (HEK293T cells) and performed pulldown assays using an antibody to the flag tag on POL. Western blot analysis with an anti-MTase mAb revealed that the MTase protein was specifically co-immunoprecipitated with POL, providing the first evidence of a specific interaction between these domains. To further assess the role of the αA3 helix in this interaction, selected residues in this motif were mutated in the recombinant MTase and the effect on POL interaction determined by the pulldown assay. These mutations were also introduced into a WNV infectious clone (FLSDX) and the replication properties of these mutant viruses assessed. While none of the αA3 mutations had a significant effect on the MTase-POL association in pulldown assays, suggesting that these residues were not specific to the interaction, an E46L mutation completely abolished virus viability indicating a critical requirement of this residue in replication. Failure to generate compensatory mutations in POL to rescue replication, even after several passages of the transfection supernatant in Vero cells, precluded further conclusion of the role of this residue in the context of MTase-POL interactions.


Subject(s)
Methyltransferases/metabolism , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication , West Nile virus/enzymology , West Nile virus/physiology , Amino Acid Substitution , Animals , Blotting, Western , Cell Line , Centrifugation , DNA Mutational Analysis , Humans , Immunoprecipitation , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
Biochim Biophys Acta ; 1834(6): 1077-88, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23410953

ABSTRACT

Based on bioinformatics interrogation of the genome, >500 mammalian protein kinases can be clustered within seven different groups. Of these kinases, the mitogen-activated protein kinase (MAPK) family forms part of the CMGC group of serine/threonine kinases that includes extracellular signal regulated kinases (ERKs), cJun N-terminal kinases (JNKs), and p38 MAPKs. With the JNKs considered attractive targets in the treatment of pathologies including diabetes and stroke, efforts have been directed to the discovery of new JNK inhibitory molecules that can be further developed as new therapeutics. Capitalizing on our biochemical understanding of JNK, we performed in silico screens of commercially available chemical databases to identify JNK1-interacting compounds and tested their in vitro JNK inhibitory activity. With in vitro and cell culture studies, we showed that the compound, 4'-methyl-N(2)-3-pyridinyl-4,5'-bi-1,3-thiazole-2,2'-diamine (JNK Docking (JD) compound 123, but not the related compound (4'-methyl-N~2~-(6-methyl-2-pyridinyl)-4,5'-bi-1,3-thiazole-2,2'-diamine (JD124), inhibited JNK1 activity towards a range of substrates. Molecular docking, saturation transfer difference NMR experiments and enzyme kinetic analyses revealed both ATP- and substrate-competitive inhibition of JNK by JD123. In characterizing JD123 further, we noted its ATP-competitive inhibition of the related p38-γ MAPK, but not ERK1, ERK2, or p38-α, p38-ß or p38-δ. Further screening of a broad panel of kinases using 10µM JD123, identified inhibition of kinases including protein kinase Bß (PKBß/Aktß). Appropriately modified thiazole diamines, as typified by JD123, thus provide a new chemical scaffold for development of inhibitors for the JNK and p38-γ MAPKs as well as other kinases that are also potential therapeutic targets such as PKBß/Aktß.


Subject(s)
Diamines/chemistry , Diamines/pharmacology , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cell Line , Cells, Cultured , Competitive Bidding , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 8/chemistry , Mitogen-Activated Protein Kinase 8/metabolism , Models, Molecular , Molecular Docking Simulation/methods , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
J Pharm Biomed Anal ; 67-68: 129-36, 2012.
Article in English | MEDLINE | ID: mdl-22591806

ABSTRACT

Thousands of metabolites are excreted in urine, and potentially can be detected in NMR spectra. Currently, NMR spectral information for about one thousand metabolites has been deposited in publicly available sources, limiting the identification of chemical compounds that are potential biomarkers for clinical and subclinical applications. This study reports the identification of crotonyl glycine, one of the key metabolites detected by ¹H NMR as excreted in the urine of sheep after 48 h road transport and during the subsequent 72 h recovery period. This metabolite was important in separating the metabolic responses as expressed in the urine from animals undergoing shorter road transport treatments. At the time of the metabonomic analysis, the NMR signals from this metabolite were designated as unassigned as no match was found in public databases or the literature. Selected sheep urine samples containing the metabolite were resolved by reversed phase HPLC reducing the sample complexity. Subsequent ¹H NMR spectra of the collected fractions revealed that the unknown metabolite was present in a single HPLC fraction. High-resolution 1D and 2D ¹H NMR spectra of this fraction followed by mass determination of the parent ion and its fragments by nanoESI-TOF-MS/MS revealed the identity of the compound as crotonyl glycine (N-but-(E)-2-enoyl glycine). The HPLC fraction was subsequently spiked with synthetic crotonyl glycine which confirmed identification.


Subject(s)
Glycine/urine , Transportation , Animals , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Sheep
11.
Bioorg Med Chem Lett ; 22(2): 916-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22209488

ABSTRACT

Protease activated receptor 2 (PAR2) is an unusual G-protein coupled receptor in being self-activated, after pruning of the N-terminus by serine proteases like trypsin and tryptase. Short synthetic peptides corresponding to the newly exposed N-terminal hexapeptide sequence also activate PAR2 on immunoinflammatory, cancer and many normal cell types. (1)H nuclear magnetic resonance (NMR) and circular dichroism (CD) spectroscopy were used here to search for structural clues to activating mechanisms of the hexapeptide agonists SLIGRL (rat), SLIGKV (human) and the peptidomimetic analogue, 2-furoyl-LIGRLO. Either with a free or acetyl capped N-terminus, these agonist peptides display significant propensity in aprotic (DMSO) or lipidic (water-SDS) solvents for turn-like conformations, which are predicted to be receptor-binding conformations in the transmembrane or loops region of PAR2. These motifs may be valuable for the design of small molecule PAR2 agonists and antagonists as prospective new drugs for regulating inflammatory and proliferative diseases.


Subject(s)
Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Humans , Models, Molecular , Molecular Conformation , Oligopeptides/chemistry , Receptor, PAR-2/metabolism , Structure-Activity Relationship
12.
Antiviral Res ; 84(3): 234-41, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19781577

ABSTRACT

The flaviviruses comprise a large group of related viruses, many of which pose a significant global human health threat, most notably the dengue viruses (DENV), West Nile virus (WNV) and yellow fever virus (YFV). Flaviviruses enter host cells via fusion of the viral and cellular membranes, a process mediated by the major viral envelope protein E as it undergoes a low pH induced conformational change in the endosomal compartment of the host cell. This essential entry stage in the flavivirus life cycle provides an attractive target for the development of antiviral agents. We performed an in silico docking screen of the Maybridge chemical database within a previously described ligand binding pocket in the dengue E protein structure that is thought to play a key role in the conformational transitions that lead to membrane fusion. The biological activity of selected compounds identified from this screen revealed low micromolar antiviral potency against dengue virus for two of the compounds. Our results also provide the first evidence that compounds selected to bind to this ligand binding site on the flavivirus E protein abrogate fusion activity. Interestingly, one of these compounds also has antiviral activity against both WNV (kunjin strain) and YFV.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , Flavivirus Infections/virology , Flavivirus/drug effects , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Animals , Binding Sites , Chlorocebus aethiops , Dengue Virus/chemistry , Dengue Virus/drug effects , Dengue Virus/physiology , Flavivirus/chemistry , Flavivirus/physiology , Flavivirus Infections/drug therapy , Humans , Protein Binding , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
13.
J Mol Biol ; 385(5): 1568-77, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19059417

ABSTRACT

Over the last decade, West Nile virus has spread rapidly via mosquito transmission from infected migratory birds to humans. One potential therapeutic approach to treating infection is to inhibit the virally encoded serine protease that is essential for viral replication. Here we report the crystal structure of the viral NS3 protease tethered to its essential NS2B cofactor and bound to a potent substrate-based tripeptide inhibitor, 2-naphthoyl-Lys-Lys-Arg-H (K(i)=41 nM), capped at the N-terminus by 2-naphthoyl and capped at the C-terminus by aldehyde. An important and unexpected feature of this structure is the presence of two conformations of the catalytic histidine suggesting a role for ligand stabilization of the catalytically competent His conformation. Analysis of other West Nile virus NS3 protease structures and related serine proteases supports this hypothesis, suggesting that the common catalytic mechanism involves an induced-fit mechanism.


Subject(s)
Models, Molecular , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry , West Nile virus/enzymology , Amino Acid Sequence , Catalysis , Catalytic Domain , Ligands , Molecular Sequence Data , Protein Conformation , RNA Helicases/antagonists & inhibitors , RNA Helicases/chemistry , Serine Proteinase Inhibitors/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , X-Ray Diffraction
14.
J Med Chem ; 51(18): 5714-21, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18729351

ABSTRACT

West Nile virus (WNV) has spread rapidly around the globe, efficiently crossing species from migrating birds into humans and other mammals. The viral protease NS2B-NS3 is important for WNV replication and recognizes dibasic substrate sequences common to other flaviviral proteases but different from most mammalian proteases. Potent inhibitors of WNV protease with antiviral activity have been elusive to date. We report the smallest and most potent inhibitors known for this enzyme, cationic tripeptides with nonpeptidic caps at the N-terminus and aldehyde at the C-terminus. One of these, compound 3 ( Ki = 9 nM) is stable in serum (>90% intact after 3 h, 37 degrees C), cell permeable, and shows antiviral activity (IC 50 1.6 microM) without cytotoxicity (IC 50 >400 microM), thereby validating the approach of inhibiting WNV protease to suppress WNV replication.


Subject(s)
Antiviral Agents/pharmacology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/blood , Antiviral Agents/chemistry , Cations , Models, Molecular , Protease Inhibitors/blood , Protease Inhibitors/chemistry , RNA Helicases/antagonists & inhibitors , RNA Helicases/chemistry , RNA Helicases/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Substrate Specificity , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
15.
J Gen Virol ; 89(Pt 4): 1010-1014, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18343843

ABSTRACT

The flavivirus NS2B/NS3 protease has received considerable attention as a target for the development of antiviral compounds. While substrate based inhibitors have been the primary focus to date, an approach focussing on NS2B cofactor displacement could prove to be an effective alternative. To understand better the role of the NS2B cofactor in protease activation, we conducted an alanine mutagenesis screen throughout the 42-residue central cofactor domain (NS2B(51-92)) of West Nile virus (WNV). Two sites critical for proteolytic activity were identified (NS2B(59-62) and NS2B(75-87)), where the majority of substitutions were found to significantly decrease proteolytic activity of a recombinant WNV NS2B/NS3 protease. These findings provide mechanistic insights into the structural and functional role that the cofactor may play in the substrate-bound and free protease complexes as well as providing novel sites for targeting new antiviral inhibitors.


Subject(s)
Endopeptidases/chemistry , Flavivirus Infections/virology , West Nile virus/chemistry , Amino Acid Sequence , Catalytic Domain , Coenzymes , Endopeptidases/genetics , Endopeptidases/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Structure, Tertiary , Sequence Alignment , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , West Nile Fever
16.
Eur J Med Chem ; 43(3): 513-39, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17582659

ABSTRACT

An exploratory chemical effort has been undertaken to develop a novel series of compounds as selective CB(1) agonists. It is hoped that compounds of this type will have clinical utility in pain control, and cerebral ischaemia following stroke or traumatic head injury. We report here medicinal chemistry studies directed towards the investigation of a series of 1-substituted-indole-3-oxadiazoles as potential CB(1) agonists.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Cannabinoids/metabolism , Indoles/chemical synthesis , Indoles/pharmacology , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Analgesics/chemistry , Analgesics/metabolism , Animals , Binding Sites , Drug Design , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Indoles/chemistry , Indoles/metabolism , Inhibitory Concentration 50 , Mice , Oxadiazoles/chemistry , Oxadiazoles/metabolism , Rats , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism , Substrate Specificity
17.
Protein Expr Purif ; 53(1): 87-96, 2007 May.
Article in English | MEDLINE | ID: mdl-17174105

ABSTRACT

West Nile virus is a medically significant emerging pathogen for which there is no effective antiviral therapy. The viral protease encoded by NS2B and NS3 is an attractive target for development of an inhibitor and has been the focus of numerous studies. Most have employed recombinant proteases based on an expression strategy we developed which links the essential hydrophilic cofactor domain within NS2B to the NS3 protease domain by a flexible glycine linker. However, autoproteolysis has been a significant problem associated with this construct. The recently resolved crystal structure of the cofactor bound WNV NS3 protease for example, was found to be truncated by 18 residues at its N-terminus. In this study, the autocatalytic cleavage site was identified and removed along with nonessential regions of the glycine linker and cofactor domain. In addition, the optimal size of the NS3 protease was defined. Based on this optimized construct, a recombinant protease incorporating the full length of NS3 was also successfully expressed and purified. Somewhat surprisingly, comparative analysis of the proteolytic activity of this recombinant with that of the protease domain alone revealed little influence of the C-terminal two thirds of NS3 on substrate binding. These modifications have yielded highly stable and constrained recombinant proteases, which are more suitable than existing constructs for both activity and structural studies.


Subject(s)
Endopeptidases/chemistry , Viral Nonstructural Proteins/chemistry , West Nile virus/enzymology , Amino Acid Sequence , Binding Sites , Chromatography, High Pressure Liquid , Enzyme Stability , Escherichia coli/genetics , Escherichia coli/metabolism , Glycine/chemistry , Histidine/chemistry , Kinetics , Mass Spectrometry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Transformation, Genetic , West Nile virus/chemistry
18.
J Biol Chem ; 281(50): 38448-58, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17052977

ABSTRACT

West Nile Virus is becoming a widespread pathogen, infecting people on at least four continents with no effective treatment for these infections or many of their associated pathologies. A key enzyme that is essential for viral replication is the viral protease NS2B-NS3, which is highly conserved among all flaviviruses. Using a combination of molecular fitting of substrates to the active site of the crystal structure of NS3, site-directed enzyme and cofactor mutagenesis, and kinetic studies on proteolytic processing of panels of short peptide substrates, we have identified important enzyme-substrate interactions that define substrate specificity for NS3 protease. In addition to better understanding the involvement of S2, S3, and S4 enzyme residues in substrate binding, a residue within cofactor NS2B has been found to strongly influence the preference of flavivirus proteases for lysine or arginine at P2 in substrates. Optimization of tetrapeptide substrates for enhanced protease affinity and processing efficiency has also provided important clues for developing inhibitors of West Nile Virus infection.


Subject(s)
Viral Nonstructural Proteins/metabolism , West Nile virus/enzymology , Amino Acid Sequence , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA Helicases/chemistry , RNA Helicases/genetics , RNA Helicases/metabolism , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Substrate Specificity , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
19.
Med Chem ; 2(1): 89-112, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16787359

ABSTRACT

In silico virtual screening for drug discovery has become a hot topic in medicinal chemistry research during the last 5 years, growing from a largely academic pursuit concerned principally with validating the methods used, to a major early-stage technique for lead discovery in the pharmaceutical industry. In this review we highlight a few recent successes in ligand docking associated with virtual screening, paying particular attention to four major target classes of pharmaceutical interest (G Protein-Coupled receptors, nuclear hormone receptors, kinases, proteases). We also discuss some emerging trends in the field, some common limitations, and how they are being overcome.


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Technology, Pharmaceutical/methods , Binding Sites , Computational Biology , Drug Design , Ligands , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Phosphotransferases/chemistry , Phosphotransferases/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , Technology, Pharmaceutical/trends
20.
Antimicrob Agents Chemother ; 50(2): 639-48, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436721

ABSTRACT

Parasite resistance to antimalarial drugs is a serious threat to human health, and novel agents that act on enzymes essential for parasite metabolism, such as proteases, are attractive targets for drug development. Recent studies have shown that clinically utilized human immunodeficiency virus (HIV) protease inhibitors can inhibit the in vitro growth of Plasmodium falciparum at or below concentrations found in human plasma after oral drug administration. The most potent in vitro antimalarial effects have been obtained for parasites treated with saquinavir, ritonavir, or lopinavir, findings confirmed in this study for a genetically distinct P. falciparum line (3D7). To investigate the potential in vivo activity of antiretroviral protease inhibitors (ARPIs) against malaria, we examined the effect of ARPI combinations in a murine model of malaria. In mice infected with Plasmodium chabaudi AS and treated orally with ritonavir-saquinavir or ritonavir-lopinavir, a delay in patency and a significant attenuation of parasitemia were observed. Using modeling and ligand docking studies we examined putative ligand binding sites of ARPIs in aspartyl proteases of P. falciparum (plasmepsins II and IV) and P. chabaudi (plasmepsin) and found that these in silico analyses support the antimalarial activity hypothesized to be mediated through inhibition of these enzymes. In addition, in vitro enzyme assays demonstrated that P. falciparum plasmepsins II and IV are both inhibited by the ARPIs saquinavir, ritonavir, and lopinavir. The combined results suggest that ARPIs have useful antimalarial activity that may be especially relevant in geographical regions where HIV and P. falciparum infections are both endemic.


Subject(s)
HIV Protease Inhibitors/pharmacology , Malaria/drug therapy , Plasmodium falciparum/drug effects , Animals , Aspartic Acid Endopeptidases/chemistry , Binding Sites , Crystallization , Female , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/therapeutic use , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Protein Conformation , Protozoan Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...