Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Negl Trop Dis ; 17(4): e0011303, 2023 04.
Article in English | MEDLINE | ID: mdl-37104530

ABSTRACT

Lymphatic filariasis is a debilitating illness with an estimated 50 million cases as of 2018. The majority of cases are caused by the parasitic worm W. bancrofti and additional cases by the worms B. malayi and B. timori. Dihydrofolate reductase (DHFR) is an established target in the treatment of cancer, bacterial, and protozoal infections and may be a potential target for drugs targeting parasitic worm infections, including filariasis. Recent studies have shown that known antifolate compounds, including methotrexate, inhibit the activity of W. bancrofti DHFR (WbDHFR). However, the absence of structural information for filarial DHFRs has limited the study of more in-depth structure-function relationships. We report the structure of WbDHFR complexed with NADPH and folate using X-ray diffraction data measured to 2.47 Å resolution. The structure of WbDHFR reveals the usual DHFR fold and is currently only the second nematode DHFR structure in the Protein Data Bank. The equilibrium dissociation constants for NADPH (90 ± 29 nM) and folate (23 ± 4 nM) were determined by equilibrium titrations. The interactions of known antifolates with WbDHFR were analyzed using molecular docking programs and molecular dynamics simulations. Antifolates with a hydrophobic core and extended linker formed favorable interactions with WbDHFR. These combined data should now facilitate the rational design of filarial DHFR inhibitors, which in turn can be used to determine whether DHFR is a viable drug target for filariasis and whether existing antifolates may be repurposed for its treatment.


Subject(s)
Elephantiasis, Filarial , Folic Acid Antagonists , Animals , Folic Acid Antagonists/pharmacology , Folic Acid Antagonists/metabolism , Wuchereria bancrofti , Folic Acid , Tetrahydrofolate Dehydrogenase/metabolism , NADP , Molecular Docking Simulation
2.
Arch Biochem Biophys ; 692: 108545, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32810476

ABSTRACT

Many antibacterial and antiparasitic drugs work by competitively inhibiting dihydrofolate reductase (DHFR), a vital enzyme in folate metabolism. The interactions between inhibitors and DHFR active site residues are known in many homologs but the contributions from distal residues are less understood. Identifying distal residues that aid in inhibitor binding can improve targeted drug development programs by accounting for distant influences that may be less conserved and subject to frequent resistance causing mutations. Previously, a novel, homology-based, computational approach that mines ligand inhibition data was used to predict residues involved in inhibitor selectivity in the DHFR family. Expectedly, some inhibitor selectivity determining residue positions were predicted to lie in the active site and coincide with experimentally known inhibitor selectivity determining positions. However, other residues that group spatially in clusters distal to the active site have not been previously investigated. In this study, the effect of introducing amino acid substitutions at one of these predicted clusters (His38-Ala39-Ile40) on the inhibitor selectivity profile in Bacillus stearothermophilus dihydrofolate reductase (Bs DHFR) was investigated. Mutations were introduced into these cluster positions to change sidechain chemistry and size. We determined kcat and KM values and measured KD values at equilibrium for two competitive DHFR inhibitors, trimethoprim (TMP) and pyrimethamine (PYR). Mutations in the His38-Ala39-Ile40 cluster significantly impacted inhibitor binding and TMP/PYR selectivity - seven out of nine mutations resulted in tighter binding to PYR when compared to TMP. These data suggest that the His38-Ala39-Ile40 cluster is a distal inhibitor selectivity determining region that favors PYR binding in Bs DHFR and, possibly, throughout the DHFR family.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Folic Acid Antagonists/chemistry , Geobacillus stearothermophilus/enzymology , Mutation, Missense , Tetrahydrofolate Dehydrogenase/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Geobacillus stearothermophilus/genetics , Tetrahydrofolate Dehydrogenase/genetics
3.
PLoS One ; 13(5): e0197173, 2018.
Article in English | MEDLINE | ID: mdl-29787565

ABSTRACT

Filariasis is a tropical disease caused by the parasitic nematodes Wuchereria bancrofti and Brugia malayi. Known inhibitors of dihydrofolate reductase (DHFR) have been previously shown to kill Brugia malayi nematodes and to inhibit Brugia malayi DHFR (BmDHFR) at nanomolar concentrations. These data suggest that BmDHFR is a potential target for the treatment of filariasis. Here, protocols for cloning, expression and purification of Wuchereria bancrofti DHFR (WbDHFR) were developed. The Uniprot entry J9F199-1 predicts a 172 amino acid protein for WbDHFR but alignment of this sequence to the previously described BmDHFR shows that this WbDHFR sequence lacks a crucial, conserved 13 amino acid loop. The presence of the loop in WbDHFR is supported by a noncanonical splicing event and the loop sequence was therefore included in the gene design. Subsequently, the KM for dihydrofolate (3.7 ± 2 µM), kcat (7.4 ± 0.6 s-1), and pH dependence of activity were determined. IC50 values of methotrexate, trimethoprim, pyrimethamine, raltitrexed, aminopterin, (-)-epicatechin gallate, (-)-epicatechin, and vitexin were measured for WbDHFR and BmDHFR. Methotrexate and structurally related aminopterin were found to be effective inhibitors of WbDHFR, with an KI of 1.2 ± 0.2 nM and 2.1 ± 0.5 nM, respectively, suggesting that repurposing of known antifolate compound may be an effective strategy to treating filariasis. Most compounds showed similar inhibition profiles toward both enzymes, suggesting that the two enzymes have important similarities in their active site environments and can be targeted with the same compound, once a successful inhibitor is identified.


Subject(s)
Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Wuchereria bancrofti/enzymology , Amino Acid Sequence , Animals , Brugia malayi/enzymology , Brugia malayi/genetics , Cloning, Molecular , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Sequence Alignment , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/isolation & purification , Wuchereria bancrofti/genetics
4.
Protein Expr Purif ; 128: 81-5, 2016 12.
Article in English | MEDLINE | ID: mdl-27544923

ABSTRACT

Brugia malayi (B. malayi) is one of the three causative agents of lymphatic filariasis, a neglected parasitic disease. Current literature suggests that dihydrofolate reductase is a potential drug target for the elimination of B. malayi. Here we report the recombinant expression and purification of a ∼20 kDa B. malayi dihydrofolate reductase (BmDHFR). A His6-tagged construct was expressed in E. coli and purified by affinity chromatography to yield active and homogeneous enzyme for steady-state kinetic characterization and inhibition studies. The catalytic activity kcat was found to be 1.4 ± 0.1 s(-1), the Michaelis Menten constant KM for dihydrofolate 14.7 ± 3.6 µM, and the equilibrium dissociation constant KD for NADPH 25 ± 24 nM. For BmDHFR, IC50 values for a six DHFR inhibitors were determined to be 3.1 ± 0.2 nM for methotrexate, 32 ± 22 µM for trimethoprim, 109 ± 34 µM for pyrimethamine, 154 ± 46 µM for 2,4-diaminoquinazoline, 771 ± 44 µM for cycloguanil, and >20,000 µM for 2,4-diaminopyrimidine. Our findings suggest that antifolate compounds can serve as inhibitors of BmDHFR.


Subject(s)
Brugia malayi/genetics , Gene Expression , Helminth Proteins , Tetrahydrofolate Dehydrogenase , Animals , Brugia malayi/enzymology , Catalysis , Helminth Proteins/biosynthesis , Helminth Proteins/chemistry , Helminth Proteins/genetics , Helminth Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Tetrahydrofolate Dehydrogenase/biosynthesis , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/isolation & purification
5.
Biochem J ; 442(3): 483-94, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22132769

ABSTRACT

Hepsin is a type II transmembrane serine protease that is expressed in several human tissues. Overexpression of hepsin has been found to correlate with tumour progression and metastasis, which is so far best studied for prostate cancer, where more than 90% of such tumours show this characteristic. To enable improved future patient treatment, we have developed a monoclonal humanized antibody that selectively inhibits human hepsin and does not inhibit other related proteases. We found that our antibody, hH35, potently inhibits hepsin enzymatic activity at nanomolar concentrations. Kinetic characterization revealed non-linear, slow, tight-binding inhibition. This correlates with the crystal structure we obtained for the human hepsin-hH35 antibody Fab fragment complex, which showed that the antibody binds hepsin around α3-helix, located far from the active centre. The unique allosteric mode of inhibition of hH35 is distinct from the recently described HGFA (hepatocyte growth factor activator) allosteric antibody inhibition. We further explain how a small change in the antibody design induces dramatic structural rearrangements in the hepsin antigen upon binding, leading to complete enzyme inactivation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Crystallography, X-Ray , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Protein Conformation , Serine Proteinase Inhibitors/chemistry , Transfection
6.
J Med Chem ; 53(9): 3502-16, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20380377

ABSTRACT

The inhibition of LTB(4) binding to and activation of G-protein-coupled receptors BLT1 and BLT2 is the premise of a treatment for several inflammatory diseases. In a lead optimization effort starting with the leukotriene B(4) (LTB(4)) receptor antagonist (2), members of a series of 3,5-diarylphenyl ethers were found to be highly potent inhibitors of LTB(4) binding to BLT1 and BLT2 receptors, with varying levels of selectivity depending on the substitution. In addition, compounds 33 and 38 from this series have good in vitro ADME properties, good oral bioavailability, and efficacy after oral delivery in guinea pig LTB(4) and nonhuman primate allergen challenge models. Further profiling in a rat non-GLP toxicity experiment provided the rationale for differentiation and selection of one compound (33) for clinical development.


Subject(s)
Drug Discovery , Leukotriene Antagonists/chemistry , Phenyl Ethers/pharmacology , Receptors, Leukotriene B4/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical , Guinea Pigs , HL-60 Cells , Humans , Leukotriene Antagonists/pharmacology , Phenyl Ethers/chemistry , Primates , Protein Binding , Rats , Receptors, G-Protein-Coupled/metabolism , Receptors, Leukotriene B4/metabolism , Structure-Activity Relationship
7.
J Exp Med ; 207(3): 591-605, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20212068

ABSTRACT

RNA splicing is an increasingly recognized regulator of immunity. Here, we demonstrate that after Mycobacterium tuberculosis infection (mRNA) il12rb1 is spliced by dendritic cells (DCs) to form an alternative (mRNA) il12rb1Deltatm that encodes the protein IL-12Rbeta1DeltaTM. Compared with IL-12Rbeta1, IL-12Rbeta1DeltaTM contains an altered C-terminal sequence and lacks a transmembrane domain. Expression of IL-12Rbeta1DeltaTM occurs in CD11c(+) cells in the lungs during M. tuberculosis infection. Selective reconstitution of il12rb1(-/-) DCs with (mRNA) il12rb1 and/or (mRNA) il12rb1Deltatm demonstrates that IL-12Rbeta1DeltaTM augments IL-12Rbeta1-dependent DC migration and activation of M. tuberculosis-specific T cells. It cannot mediate these activities independently of IL12Rbeta1. We hypothesize that M. tuberculosis-exposed DCs express IL-12Rbeta1DeltaTM to enhance IL-12Rbeta1-dependent migration and promote M. tuberculosis-specific T cell activation. IL-12Rbeta1DeltaTM thus represents a novel positive-regulator of IL12Rbeta1-dependent DC function and of the immune response to M. tuberculosis.


Subject(s)
Dendritic Cells/immunology , Mycobacterium tuberculosis/genetics , Receptors, Interleukin-12/genetics , Alternative Splicing , Animals , Bone Marrow Cells/physiology , Cell Movement , Chemokine CCL19/physiology , Dendritic Cells/physiology , Kinetics , Lung/immunology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/immunology , RNA, Messenger/genetics , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/immunology
8.
Prostaglandins Other Lipid Mediat ; 92(1-4): 33-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20214997

ABSTRACT

Asthma, chronic obstructive pulmonary disease (COPD) and acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are characterized by neutrophilic inflammation and elevated levels of leukotriene B4 (LTB4). However, the exact role of LTB4 pathways in mediating pulmonary neutrophilia and the potential therapeutic application of LTB4 receptor antagonists in these diseases remains controversial. Here we show that a novel dual BLT1 and BLT2 receptor antagonist, RO5101576, potently inhibited LTB4-evoked calcium mobilization in HL-60 cells and chemotaxis of human neutrophils. RO5101576 significantly attenuated LTB4-evoked pulmonary eosinophilia in guinea pigs. In non-human primates, RO5101576 inhibited allergen and ozone-evoked pulmonary neutrophilia, with comparable efficacy to budesonide (allergic responses). RO5101576 had no effects on LPS-evoked neutrophilia in guinea pigs and cigarette smoke-evoked neutrophilia in mice and rats. In toxicology studies RO5101576 was well-tolerated. Theses studies show differential effects of LTB4 receptor antagonism on neutrophil responses in vivo and suggest RO5101576 may represent a potential new treatment for pulmonary neutrophilia in asthma.


Subject(s)
Benzodioxoles/pharmacology , Phenylpropionates/pharmacology , Pneumonia/drug therapy , Primates , Receptors, Leukotriene B4/antagonists & inhibitors , Animals , Benzodioxoles/therapeutic use , Benzodioxoles/toxicity , Dogs , Drug-Related Side Effects and Adverse Reactions , Female , Guinea Pigs , HL-60 Cells , Humans , Hypersensitivity/complications , Lipopolysaccharides/pharmacology , Lung/drug effects , Male , Mice , Ozone/pharmacology , Phenylpropionates/therapeutic use , Phenylpropionates/toxicity , Pneumonia/chemically induced , Pneumonia/complications , Pneumonia/metabolism , Rats , Receptors, Leukotriene B4/metabolism , Smoking/adverse effects , Toxicity Tests
9.
J Biomol Screen ; 10(8): 832-40, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16234341

ABSTRACT

Several commercially available pharmaceutical compounds have been shown to block the IKr current of the cardiac action potential. This effect can cause a prolongation of the electrocardiogram QT interval and a delay in ventricular repolarization. The Food and Drug Administration recommends that all new potential drug candidates be assessed for IKr block to avoid a potentially lethal cardiac arrhythmia known as torsades de pointes. Direct compound interaction with the human ether-a-go-go- related gene (hERG) product, a delayed rectifier potassium channel, has been identified as a molecular mechanism of IKr block. One strategy to identify compounds with hERG liability is to monitor hERG current inhibition using electrophysiology techniques. The authors describe the Ion Works HT instrument as a tool for screening cell lines expressing hERG channels. Based on current amplitude and stability criteria, a cell line was selected and used to perform a 300-compound screen. The screen was able to identify compounds with hERG activity within projects that spanned different therapeutic areas. The cell line selection and optimization, as well as the screening abilities of the Ion Works HT system, provide a powerful means of assessing hERGactive compounds early in the drug discovery pipeline.


Subject(s)
Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions , Electrophysiology , Ether-A-Go-Go Potassium Channels/metabolism , Action Potentials/drug effects , Animals , CHO Cells/metabolism , Cell Line , Cricetinae , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Humans , Inhibitory Concentration 50 , Long QT Syndrome/chemically induced , Patch-Clamp Techniques , Torsades de Pointes/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...