Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(9): 7443-7457, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38683753

ABSTRACT

Acanthamoeba are free-living pathogenic protozoa that cause blinding keratitis, disseminated infection, and granulomatous amebic encephalitis, which is generally fatal. The development of efficient and safe drugs is a critical unmet need. Acanthamoeba sterol 14α-demethylase (CYP51) is an essential enzyme of the sterol biosynthetic pathway. Repurposing antifungal azoles for amoebic infections has been reported, but their inhibitory effects on Acanthamoeba CYP51 enzymatic activity have not been studied. Here, we report catalytic properties, inhibition, and structural characterization of CYP51 from Acanthamoeba castellanii. The enzyme displays a 100-fold substrate preference for obtusifoliol over lanosterol, supporting the plant-like cycloartenol-based pathway in the pathogen. The strongest inhibition was observed with voriconazole (1 h IC50 0.45 µM), VT1598 (0.25 µM), and VT1161 (0.20 µM). The crystal structures of A. castellanii CYP51 with bound VT1161 (2.24 Å) and without an inhibitor (1.95 Å), presented here, can be used in the development of azole-based scaffolds to achieve optimal amoebicidal effectiveness.


Subject(s)
14-alpha Demethylase Inhibitors , Sterol 14-Demethylase , Sterol 14-Demethylase/metabolism , Sterol 14-Demethylase/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , 14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/chemical synthesis , Structure-Activity Relationship , Acanthamoeba/enzymology , Acanthamoeba/drug effects , Acanthamoeba castellanii/enzymology , Acanthamoeba castellanii/drug effects , Crystallography, X-Ray , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/chemical synthesis , Models, Molecular , Molecular Structure
2.
Angew Chem Int Ed Engl ; 63(9): e202317711, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38206808

ABSTRACT

The 14α-demethylation step is critical in eukaryotic sterol biosynthesis, catalyzed by cytochrome P450 (P450) Family 51 enzymes, for example, with lanosterol in mammals. This conserved three-step reaction terminates in a C-C cleavage step that generates formic acid, the nature of which has been controversial. Proposed mechanisms involve roles of P450 Compound 0 (ferric peroxide anion, FeO2 - ) or Compound I (perferryl oxygen, FeO3+ ) reacting with either the aldehyde or its hydrate, respectively. Analysis of 18 O incorporation into formic acid from 18 O2 provides a means of distinguishing the two mechanisms. Human P450 51A1 incorporated 88 % 18 O (one atom) into formic acid, consistent with a major but not exclusive FeO2 - mechanism. Two P450 51 orthologs from amoeba and yeast showed similar results, while two orthologs from pathogenic trypanosomes showed roughly equal contributions of both mechanisms. An X-ray crystal structure of the human enzyme showed the aldehyde oxygen atom 3.5 Šaway from the heme iron atom. Experiments with human P450 51A1 and H2 18 O yielded primarily one 18 O atom but 14 % of the formic acid product with two 18 O atoms, indicative of a minor contribution of a Compound I mechanism. LC-MS evidence for a Compound 0-derived Baeyer-Villiger reaction product (a 14α-formyl ester) was also found.


Subject(s)
Cytochrome P-450 Enzyme System , Formates , Oxygen Isotopes , Sterols , Animals , Humans , Cytochrome P-450 Enzyme System/metabolism , Oxygen/chemistry , Saccharomyces cerevisiae/metabolism , Aldehydes , Demethylation , Mammals/metabolism
3.
J Biol Chem ; 299(7): 104841, 2023 07.
Article in English | MEDLINE | ID: mdl-37209823

ABSTRACT

Cytochrome P450 (P450, CYP) family 51 enzymes catalyze the 14α-demethylation of sterols, leading to critical products used for membranes and the production of steroids, as well as signaling molecules. In mammals, P450 51 catalyzes the 3-step, 6-electron oxidation of lanosterol to form (4ß,5α)-4,4-dimethyl-cholestra-8,14,24-trien-3-ol (FF-MAS). P450 51A1 can also use 24,25-dihydrolanosterol (a natural substrate in the Kandutsch-Russell cholesterol pathway). 24,25-Dihydrolanosterol and the corresponding P450 51A1 reaction intermediates, the 14α-alcohol and -aldehyde derivatives of dihydrolanosterol, were synthesized to study the kinetic processivity of the overall 14α-demethylation reaction of human P450 51A1. A combination of steady-state kinetic parameters, steady-state binding constants, dissociation rates of P450-sterol complexes, and kinetic modeling of the time course of oxidation of a P450-dihydrolanosterol complex showed that the overall reaction is highly processive, with koff rates of P450 51A1-dihydrolanosterol and the 14α-alcohol and 14α-aldehyde complexes being 1 to 2 orders of magnitude less than the forward rates of competing oxidations. epi-Dihydrolanosterol (the 3α-hydroxy analog) was as efficient as the common 3ß-hydroxy isomer in the binding and formation of dihydro FF-MAS. The common lanosterol contaminant dihydroagnosterol was found to be a substrate of human P450 51A1, with roughly one-half the activity of dihydrolanosterol. Steady-state experiments with 14α-methyl deuterated dihydrolanosterol showed no kinetic isotope effect, indicating that C-14α C-H bond breaking is not rate-limiting in any of the individual steps. The high processivity of this reaction generates higher efficiency and also renders the reaction less sensitive to inhibitors.


Subject(s)
Cytochrome P-450 Enzyme System , Demethylation , Lanosterol , Humans , Catalysis , Cytochrome P-450 Enzyme System/metabolism , Kinetics , Lanosterol/chemistry , Lanosterol/metabolism , Oxidation-Reduction
4.
Sci Rep ; 12(1): 16232, 2022 09 28.
Article in English | MEDLINE | ID: mdl-36171457

ABSTRACT

The molecular evolution of cytochromes P450 and associated redox-driven oxidative catalysis remains a mystery in biology. It is widely believed that sterol 14α-demethylase (CYP51), an essential enzyme of sterol biosynthesis, is the ancestor of the whole P450 superfamily given its conservation across species in different biological kingdoms. Herein we have utilized X-ray crystallography, molecular dynamics simulations, phylogenetics and electron transfer measurements to interrogate the nature of P450-redox partner binding using the naturally occurring fusion protein, CYP51-ferredoxin found in the sterol-producing bacterium Methylococcus capsulatus. Our data advocates that the electron transfer mechanics in the M. capsulatus CYP51-ferredoxin fusion protein involves an ensemble of ferredoxin molecules in various orientations and the interactions are transient. Close proximity of ferredoxin, however, is required to complete the substrate-induced large-scale structural switch in the P450 domain that enables proton-coupled electron transfer and subsequent oxygen scission and catalysis. These results have fundamental implications regarding the early evolution of electron transfer proteins and for the redox reactions in the early steps of sterol biosynthesis. They also shed new light on redox protein mechanics and the subsequent diversification of the P450 electron transfer machinery in nature.


Subject(s)
Ferredoxins , Protons , Cytochrome P-450 Enzyme System/metabolism , Electrons , Ferredoxins/metabolism , Oxidation-Reduction , Oxygen/metabolism , Sterol 14-Demethylase/chemistry , Sterols
5.
J Med Chem ; 64(23): 17511-17522, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34842434

ABSTRACT

Naegleria fowleri is the protozoan pathogen that causes primary amoebic meningoencephalitis (PAM), with the death rate exceeding 97%. The amoeba makes sterols and can be targeted by sterol biosynthesis inhibitors. Here, we characterized N. fowleri sterol 14-demethylase, including catalytic properties and inhibition by clinical antifungal drugs and experimental substituted azoles with favorable pharmacokinetics and low toxicity. None of them inhibited the enzyme stoichiometrically. The highest potencies were displayed by posaconazole (IC50 = 0.69 µM) and two of our compounds (IC50 = 1.3 and 0.35 µM). Because both these compounds penetrate the brain with concentrations reaching minimal inhibitory concentration (MIC) values in an N. fowleri cellular assay, we report them as potential drug candidates for PAM. The 2.1 Å crystal structure, in complex with the strongest inhibitor, provides an explanation connecting the enzyme weaker substrate specificity with lower sensitivity to inhibition. It also provides insight into the enzyme/ligand molecular recognition process and suggests directions for the design of more potent inhibitors.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Naegleria fowleri/enzymology , Sterol 14-Demethylase/metabolism , Ligands , Sterol 14-Demethylase/drug effects , Substrate Specificity
6.
Mol Biol Evol ; 38(3): 952-967, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33031537

ABSTRACT

Sterol biosynthesis, primarily associated with eukaryotic kingdoms of life, occurs as an abbreviated pathway in the bacterium Methylococcus capsulatus. Sterol 14α-demethylation is an essential step in this pathway and is catalyzed by cytochrome P450 51 (CYP51). In M. capsulatus, the enzyme consists of the P450 domain naturally fused to a ferredoxin domain at the C-terminus (CYP51fx). The structure of M. capsulatus CYP51fx was solved to 2.7 Å resolution and is the first structure of a bacterial sterol biosynthetic enzyme. The structure contained one P450 molecule per asymmetric unit with no electron density seen for ferredoxin. We connect this with the requirement of P450 substrate binding in order to activate productive ferredoxin binding. Further, the structure of the P450 domain with bound detergent (which replaced the substrate upon crystallization) was solved to 2.4 Å resolution. Comparison of these two structures to the CYP51s from human, fungi, and protozoa reveals strict conservation of the overall protein architecture. However, the structure of an "orphan" P450 from nonsterol-producing Mycobacterium tuberculosis that also has CYP51 activity reveals marked differences, suggesting that loss of function in vivo might have led to alterations in the structural constraints. Our results are consistent with the idea that eukaryotic and bacterial CYP51s evolved from a common cenancestor and that early eukaryotes may have recruited CYP51 from a bacterial source. The idea is supported by bioinformatic analysis, revealing the presence of CYP51 genes in >1,000 bacteria from nine different phyla, >50 of them being natural CYP51fx fusion proteins.


Subject(s)
Evolution, Molecular , Methylococcus capsulatus/genetics , Sterol 14-Demethylase/genetics , Animals , Humans , Methylococcus capsulatus/enzymology , Protein Conformation , Sterol 14-Demethylase/chemistry
7.
J Biol Chem ; 295(29): 9998-10007, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32493730

ABSTRACT

CYP51 enzymes (sterol 14α-demethylases) are cytochromes P450 that catalyze multistep reactions. The CYP51 reaction occurs in all biological kingdoms and is essential in sterol biosynthesis. It removes the 14α-methyl group from cyclized sterol precursors by first forming an alcohol, then an aldehyde, and finally eliminating formic acid with the introduction of a Δ14-15 double bond in the sterol core. The first two steps are typical hydroxylations, mediated by an electrophilic compound I mechanism. The third step, C-C bond cleavage, has been proposed to involve either compound I (i.e. FeO3+) or, alternatively, a proton transfer-independent nucleophilic ferric peroxo anion (compound 0, i.e. Fe3+O2-). Here, using comparative crystallographic and biochemical analyses of WT human CYP51 (CYP51A1) and its D231A/H314A mutant, whose proton delivery network is destroyed (as evidenced in a 1.98-Å X-ray structure in complex with lanosterol), we demonstrate that deformylation of the 14α-carboxaldehyde intermediate requires an active proton relay network to drive the catalysis. These results indicate a unified, compound I-based mechanism for all three steps of the CYP51 reaction, as previously established for CYP11A1 and CYP19A1. We anticipate that our approach can be applied to mechanistic studies of other P450s that catalyze multistep reactions, such as C-C bond cleavage.


Subject(s)
Protons , Sterol 14-Demethylase/chemistry , Aromatase/chemistry , Catalysis , Cholesterol Side-Chain Cleavage Enzyme/chemistry , Crystallography, X-Ray , Humans
8.
J Med Chem ; 62(22): 10391-10401, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31663733

ABSTRACT

Sterol 14α-demethylases (CYP51) are the cytochrome P450 enzymes required for biosynthesis of sterols in eukaryotes, the major targets for antifungal agents and prospective targets for treatment of protozoan infections. Human CYP51 could be and, for a while, was considered as a potential target for cholesterol-lowering drugs (the role that is now played by statins, which are also in clinical trials for cancer) but revealed high intrinsic resistance to inhibition. While microbial CYP51 enzymes are often inhibited stoichiometrically and functionally irreversibly, no strong inhibitors have been identified for human CYP51. In this study, we used comparative structure/functional analysis of CYP51 orthologs from different biological kingdoms and employed site-directed mutagenesis to elucidate the molecular basis for the resistance of the human enzyme to inhibition and also designed, synthesized, and characterized new compounds. Two of them inhibit human CYP51 functionally irreversibly with their potency approaching the potencies of azole drugs currently used to inhibit microbial CYP51.


Subject(s)
14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , Sterol 14-Demethylase/chemistry , Sterol 14-Demethylase/genetics , 14-alpha Demethylase Inhibitors/chemical synthesis , Animals , Catalytic Domain , Crystallography, X-Ray , Drug Design , Humans , Molecular Structure , Mutagenesis, Site-Directed , Protozoan Proteins/antagonists & inhibitors , Sterol 14-Demethylase/metabolism
9.
J Med Chem ; 61(23): 10910-10921, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30451500

ABSTRACT

Sterol 14α-demethylases (CYP51) are cytochrome P450 enzymes essential for sterol biosynthesis in eukaryotes and therapeutic targets for antifungal azoles. Multiple attempts to repurpose antifungals for treatment of human infections with protozoa (Trypanosomatidae) have been undertaken, yet so far none of them have revealed sufficient efficacy. VNI and its derivative VFV are two potent experimental inhibitors of Trypanosomatidae CYP51, effective in vivo against Chagas disease, visceral leishmaniasis, and sleeping sickness and currently under consideration as antiprotozoal drug candidates. However, VNI is less potent against Leishmania and drug-resistant strains of Trypanosoma cruzi and VFV, while displaying a broader spectrum of antiprotozoal activity, and is metabolically less stable. In this work we have designed, synthesized, and characterized a set of close analogues and identified two new compounds (7 and 9) that exceed VNI/VFV in their spectra of antiprotozoal activity, microsomal stability, and pharmacokinetics (tissue distribution in particular) and, like VNI/VFV, reveal no acute toxicity.


Subject(s)
14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , Chagas Disease/drug therapy , Drug Design , Sterol 14-Demethylase/metabolism , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/physiology , 14-alpha Demethylase Inhibitors/metabolism , 14-alpha Demethylase Inhibitors/therapeutic use , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Drug Stability , Humans , Microsomes/metabolism , Models, Molecular , Protein Conformation , Sterol 14-Demethylase/chemistry
10.
J Biol Chem ; 293(50): 19344-19353, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30327430

ABSTRACT

Sterol 14α-demethylases (CYP51s) are phylogenetically the most conserved cytochromes P450, and their three-step reaction is crucial for biosynthesis of sterols and serves as a leading target for clinical and agricultural antifungal agents. The structures of several (bacterial, protozoan, fungal, and human) CYP51 orthologs, in both the ligand-free and inhibitor-bound forms, have been determined and have revealed striking similarity at the secondary and tertiary structural levels, despite having low sequence identity. Moreover, in contrast to many of the substrate-promiscuous, drug-metabolizing P450s, CYP51 structures do not display substantial rearrangements in their backbones upon binding of various inhibitory ligands, essentially representing a snapshot of the ligand-free sterol 14α-demethylase. Here, using the obtusifoliol-bound I105F variant of Trypanosoma cruzi CYP51, we report that formation of the catalytically competent complex with the physiological substrate triggers a large-scale conformational switch, dramatically reshaping the enzyme active site (3.5-6.0 Å movements in the FG arm, HI arm, and helix C) in the direction of catalysis. Notably, our X-ray structural analyses revealed that the substrate channel closes, the proton delivery route opens, and the topology and electrostatic potential of the proximal surface reorganize to favor interaction with the electron-donating flavoprotein partner, NADPH-cytochrome P450 reductase. Site-directed mutagenesis of the amino acid residues involved in these events revealed a key role of active-site salt bridges in contributing to the structural dynamics that accompanies CYP51 function. Comparative analysis of apo-CYP51 and its sterol-bound complex provided key conceptual insights into the molecular mechanisms of CYP51 catalysis, functional conservation, lineage-specific substrate complementarity, and druggability differences.


Subject(s)
Sterol 14-Demethylase/chemistry , Sterol 14-Demethylase/metabolism , Biocatalysis , Electron Transport , Enzyme Stability , Heme/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Trypanosoma cruzi/enzymology
11.
J Med Chem ; 61(13): 5679-5691, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29894182

ABSTRACT

Because of the increase in the number of immunocompromised patients, the incidence of invasive fungal infections is growing, but the treatment efficiency remains unacceptably low. The most potent clinical systemic antifungals (azoles) are the derivatives of two scaffolds: ketoconazole and fluconazole. Being the safest antifungal drugs, they still have shortcomings, mainly because of pharmacokinetics and resistance. Here, we report the successful use of the target fungal enzyme, sterol 14α-demethylase (CYP51), for structure-based design of novel antifungal drug candidates by minor modifications of VNI [( R)- N-(1-(2,4-dichlorophenyl)-2-(1 H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide)], an inhibitor of protozoan CYP51 that cures Chagas disease. The synthesis of fungi-oriented VNI derivatives, analysis of their potencies to inhibit CYP51s from two major fungal pathogens ( Aspergillus fumigatus and Candida albicans), microsomal stability, effects in fungal cells, and structural characterization of A. fumigatus CYP51 in complexes with the most potent compound are described, offering a new antifungal drug scaffold and outlining directions for its further optimization.


Subject(s)
Aspergillus fumigatus/drug effects , Candida albicans/drug effects , Drug Design , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Sterol 14-Demethylase/metabolism , 14-alpha Demethylase Inhibitors/chemical synthesis , 14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Aspergillus fumigatus/enzymology , Candida albicans/enzymology , Catalytic Domain , Chemistry Techniques, Synthetic , Crystallography, X-Ray , Imidazoles/chemistry , Ligands , Models, Molecular , Sterol 14-Demethylase/chemistry
12.
Article in English | MEDLINE | ID: mdl-28461309

ABSTRACT

Within the past few decades, the incidence and complexity of human fungal infections have increased, and therefore, the need for safer and more efficient, broad-spectrum antifungal agents is high. In the study described here, we characterized the new tetrazole-based drug candidate VT-1598 as an inhibitor of sterol 14α-demethylase (CYP51B) from the filamentous fungus Aspergillus fumigatus VT-1598 displayed a high affinity of binding to the enzyme in solution (dissociation constant, 13 ± 1 nM) and in the reconstituted enzymatic reaction was revealed to have an inhibitory potency stronger than the potencies of all other simultaneously tested antifungal drugs, including fluconazole, voriconazole, ketoconazole, and posaconazole. The X-ray structure of the VT-1598/A. fumigatus CYP51 complex was determined and depicts the distinctive binding mode of the inhibitor in the enzyme active site, suggesting the molecular basis of the improved drug potency and broad-spectrum antifungal activity. These data show the formation of an optimized hydrogen bond between the phenoxymethyl oxygen of VT-1598 and the imidazole ring nitrogen of His374, the CYP51 residue that is highly conserved across fungal pathogens and fungus specific. Comparative structural analysis of A. fumigatus CYP51/voriconazole and Candida albicans CYP51/VT-1161 complexes supports the role of H bonding in fungal CYP51/inhibitor complexes and emphasizes the importance of an optimal distance between this interaction and the inhibitor-heme iron interaction. Cellular experiments using two A. fumigatus strains (strains 32820 and 1022) displayed a direct correlation between the effects of the drugs on CYP51B activity and fungal growth inhibition, indicating the noteworthy anti-A. fumigatus potency of VT-1598 and confirming its promise as a broad-spectrum antifungal agent.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/enzymology , Drugs, Investigational/pharmacology , Sterol 14-Demethylase/metabolism , Aspergillus fumigatus/genetics , Candida albicans/drug effects , Candida albicans/enzymology , Candida albicans/genetics , Fluconazole/pharmacology , Ketoconazole/pharmacology , Microbial Sensitivity Tests , Pyridines/pharmacology , Sterol 14-Demethylase/genetics , Tetrazoles/pharmacology , Triazoles/pharmacology , Voriconazole/pharmacology
13.
J Biol Chem ; 292(16): 6728-6743, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28258218

ABSTRACT

With some advances in modern medicine (such as cancer chemotherapy, broad exposure to antibiotics, and immunosuppression), the incidence of opportunistic fungal pathogens such as Candida albicans has increased. Cases of drug resistance among these pathogens have become more frequent, requiring the development of new drugs and a better understanding of the targeted enzymes. Sterol 14α-demethylase (CYP51) is a cytochrome P450 enzyme required for biosynthesis of sterols in eukaryotic cells and is the major target of clinical drugs for managing fungal pathogens, but some of the CYP51 key features important for rational drug design have remained obscure. We report the catalytic properties, ligand-binding profiles, and inhibition of enzymatic activity of C. albicans CYP51 by clinical antifungal drugs that are used systemically (fluconazole, voriconazole, ketoconazole, itraconazole, and posaconazole) and topically (miconazole and clotrimazole) and by a tetrazole-based drug candidate, VT-1161 (oteseconazole: (R)-2-(2,4-difluorophenyl)-1,1-difluoro-3-(1H-tetrazol-1-yl)-1-(5-(4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)propan-2-ol). Among the compounds tested, the first-line drug fluconazole was the weakest inhibitor, whereas posaconazole and VT-1161 were the strongest CYP51 inhibitors. We determined the X-ray structures of C. albicans CYP51 complexes with posaconazole and VT-1161, providing a molecular mechanism for the potencies of these drugs, including the activity of VT-1161 against Candida krusei and Candida glabrata, pathogens that are intrinsically resistant to fluconazole. Our comparative structural analysis outlines phylum-specific CYP51 features that could direct future rational development of more efficient broad-spectrum antifungals.


Subject(s)
Antifungal Agents/chemistry , Azoles/chemistry , Candida albicans/enzymology , Fungal Proteins/chemistry , Sterol 14-Demethylase/chemistry , Sterols/biosynthesis , Animals , Crystallization , Heme/chemistry , Humans , Kinetics , Ligands , Microbial Sensitivity Tests , Protein Binding , Protein Conformation , Protons , Rats
14.
J Lipid Res ; 57(8): 1552-63, 2016 08.
Article in English | MEDLINE | ID: mdl-27313059

ABSTRACT

Rapidly multiplying cancer cells synthesize greater amounts of cholesterol to build their membranes. Cholesterol-lowering drugs (statins) are currently in clinical trials for anticancer chemotherapy. However, given at higher doses, statins cause serious side effects by inhibiting the formation of other biologically important molecules derived from mevalonate. Sterol 14α-demethylase (CYP51), which acts 10 steps downstream, is potentially a more specific drug target because this portion of the pathway is fully committed to cholesterol production. However, screening a variety of commercial and experimental inhibitors of microbial CYP51 orthologs revealed that most of them (including all clinical antifungals) weakly inhibit human CYP51 activity, even if they display high apparent spectral binding affinity. Only one relatively potent compound, (R)-N-(1-(3,4'-difluorobiphenyl-4-yl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide (VFV), was identified. VFV has been further tested in cellular experiments and found to decrease proliferation of different cancer cell types. The crystal structures of human CYP51-VFV complexes (2.0 and 2.5 Å) both display a 2:1 inhibitor/enzyme stoichiometry, provide molecular insights regarding a broader substrate profile, faster catalysis, and weaker susceptibility of human CYP51 to inhibition, and outline directions for the development of more potent inhibitors.


Subject(s)
14-alpha Demethylase Inhibitors/chemistry , Antineoplastic Agents/chemistry , Sterol 14-Demethylase/chemistry , Antifungal Agents , Antiprotozoal Agents/chemistry , Catalytic Domain , Cell Line, Tumor , Cholestadienols/chemistry , Crystallography, X-Ray , Drug Design , Drug Screening Assays, Antitumor , Humans , Hydrogen Bonding , Lanosterol/chemistry , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical
15.
Antimicrob Agents Chemother ; 60(2): 1058-66, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643331

ABSTRACT

A novel antifungal drug candidate, the 1-tetrazole-based agent VT-1161 [(R)-2-(2,4-difluorophenyl)-1,1-difluoro-3-(1H-tetrazol-1-yl)-1-{5-[4-(2,2,2-trifluoroethoxy)phenyl]pyridin-2-yl}propan-2-ol], which is currently in two phase 2b antifungal clinical trials, was found to be a tight-binding ligand (apparent dissociation constant [Kd], 24 nM) and a potent inhibitor of cytochrome P450 sterol 14α-demethylase (CYP51) from the protozoan pathogen Trypanosoma cruzi. Moreover, VT-1161 revealed a high level of antiparasitic activity against amastigotes of the Tulahuen strain of T. cruzi in cellular experiments (50% effective concentration, 2.5 nM) and was active in vivo, causing >99.8% suppression of peak parasitemia in a mouse model of infection with the naturally drug-resistant Y strain of the parasite. The data strongly support the potential utility of VT-1161 in the treatment of Chagas disease. The structural characterization of T. cruzi CYP51 in complex with VT-1161 provides insights into the molecular basis for the compound's inhibitory potency and paves the way for the further rational development of this novel, tetrazole-based inhibitory chemotype both for antiprotozoan chemotherapy and for antifungal chemotherapy.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Pyridines/pharmacology , Sterol 14-Demethylase/chemistry , Tetrazoles/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , 14-alpha Demethylase Inhibitors/chemistry , Animals , Chagas Disease/drug therapy , Crystallography, X-Ray , Disease Models, Animal , Female , Heme/chemistry , Mice , Models, Molecular , Protein Conformation , Pyridines/chemistry , Sterol 14-Demethylase/metabolism , Tetrazoles/chemistry , Trypanosoma cruzi/enzymology
16.
J Biol Chem ; 290(39): 23916-34, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26269599

ABSTRACT

Aspergillus fumigatus is the opportunistic fungal pathogen that predominantly affects the immunocompromised population and causes 600,000 deaths/year. The cytochrome P450 51 (CYP51) inhibitor voriconazole is currently the drug of choice, yet the treatment efficiency remains low, calling for rational development of more efficient agents. A. fumigatus has two CYP51 genes, CYP51A and CYP51B, which share 59% amino acid sequence identity. CYP51B is expressed constitutively, whereas gene CYP51A is reported to be inducible. We expressed, purified, and characterized A. fumigatus CYP51B, including determination of its substrate preferences, catalytic parameters, inhibition, and x-ray structure in complexes with voriconazole and the experimental inhibitor (R)-N-(1-(2,4-dichlorophenyl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide (VNI). The enzyme demethylated its natural substrate eburicol and the plant CYP51 substrate obtusifoliol at steady-state rates of 17 and 16 min(-1), respectively, but did not metabolize lanosterol, and the topical antifungal drug miconazole was the strongest inhibitor that we identified. The x-ray crystal structures displayed high overall similarity of A. fumigatus CYP51B to CYP51 orthologs from other biological kingdoms but revealed phylum-specific differences relevant to enzyme catalysis and inhibition. The complex with voriconazole provides an explanation for the potency of this relatively small molecule, whereas the complex with VNI outlines a direction for further enhancement of the efficiency of this new inhibitory scaffold to treat humans afflicted with filamentous fungal infections.


Subject(s)
Aspergillus fumigatus/enzymology , Cytochrome P-450 Enzyme System/chemistry , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/chemistry , Voriconazole/chemistry , Aspergillus fumigatus/genetics , Catalysis , Cytochrome P-450 Enzyme System/genetics , Drug Resistance, Fungal , Fungal Proteins/genetics , Protein Structure, Tertiary , Structure-Activity Relationship
17.
J Infect Dis ; 212(9): 1439-48, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-25883390

ABSTRACT

Sterol 14α-demethylases (CYP51) are the enzymes essential for sterol biosynthesis. They serve as clinical targets for antifungal azoles and are considered as targets for treatment of human Trypanosomatidae infections. Recently, we have shown that VNI, a potent and selective inhibitor of trypanosomal CYP51 that we identified and structurally characterized in complex with the enzyme, can cure the acute and chronic forms of Chagas disease. The purpose of this work was to apply the CYP51 structure/function for further development of the VNI scaffold. As anticipated, VFV (R)-N-(1-(3,4'-difluorobiphenyl-4-yl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide, the derivative designed to fill the deepest portion of the CYP51 substrate-binding cavity, reveals a broader antiprotozoan spectrum of action. It has stronger antiparasitic activity in cellular experiments, cures the experimental Chagas disease with 100% efficacy, and suppresses visceral leishmaniasis by 89% (vs 60% for VNI). Oral bioavailability, low off-target activity, favorable pharmacokinetics and tissue distribution characterize VFV as a promising new drug candidate.


Subject(s)
Antiprotozoal Agents/pharmacology , Benzamides/pharmacology , Chagas Disease/drug therapy , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/chemistry , Leishmaniasis, Visceral/drug therapy , Oxadiazoles/pharmacology , Animals , Antiprotozoal Agents/pharmacokinetics , Benzamides/pharmacokinetics , Biotransformation , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Disease Models, Animal , Female , Humans , Imidazoles/pharmacology , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Molecular Structure , Oxadiazoles/pharmacokinetics , Rats , Structure-Activity Relationship , Tissue Distribution , Trypanosoma cruzi/drug effects
18.
FEBS Lett ; 588(21): 3878-85, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25217832

ABSTRACT

CYP51 (sterol 14α-demethylase) is an efficient target for clinical and agricultural antifungals and an emerging target for treatment of Chagas disease, the infection that is caused by multiple strains of a protozoan pathogen Trypanosoma cruzi. Here, we analyze CYP51A from the Y strain T. cruzi. In this protein, proline 355, a residue highly conserved across the CYP51 family, is replaced with serine. The purified enzyme retains its catalytic activity, yet has been found less susceptible to inhibition. These biochemical data are consistent with cellular experiments, both in insect and human stages of the pathogen. Comparative structural analysis of CYP51 complexes with VNI and two derivatives suggests that broad-spectrum CYP51 inhibitors are likely to be preferable as antichagasic drug candidates.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Trypanosoma cruzi/enzymology , 14-alpha Demethylase Inhibitors/therapeutic use , Amino Acid Sequence , Amino Acid Substitution , Animals , Biocatalysis , Chagas Disease/drug therapy , Chagas Disease/enzymology , Conserved Sequence , Cytochrome P-450 Enzyme System/genetics , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Trypanosoma cruzi/drug effects
19.
J Med Chem ; 57(15): 6704-17, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25033013

ABSTRACT

Chagas disease, which was once thought to be confined to endemic regions of Latin America, has now gone global, becoming a new worldwide challenge with no cure available. The disease is caused by the protozoan parasite Trypanosoma cruzi, which depends on the production of endogenous sterols, and therefore can be blocked by sterol 14α-demethylase (CYP51) inhibitors. Here we explore the spectral binding parameters, inhibitory effects on T. cruzi CYP51 activity, and antiparasitic potencies of a new set of ß-phenyl imidazoles. Comparative structural characterization of the T. cruzi CYP51 complexes with the three most potent inhibitors reveals two opposite binding modes of the compounds ((R)-6, EC50=1.2 nM, vs (S)-2/(S)-3, EC50=1.0/5.5 nM) and suggests the entrance into the CYP51 substrate access channel and the heme propionate-supporting ceiling of the binding cavity as two distinct areas of the protein that enhance molecular recognition and therefore could be used for the development of more effective antiparasitic drugs.


Subject(s)
14-alpha Demethylase Inhibitors/chemistry , Carbamates/chemistry , Imidazoles/chemistry , Sterol 14-Demethylase/metabolism , Trypanocidal Agents/chemistry , Trypanosoma cruzi/enzymology , 14-alpha Demethylase Inhibitors/chemical synthesis , 14-alpha Demethylase Inhibitors/pharmacology , Carbamates/chemical synthesis , Carbamates/pharmacology , Crystallography, X-Ray , Drug Design , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Models, Molecular , Protein Binding , Protein Conformation , Stereoisomerism , Sterol 14-Demethylase/chemistry , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects
20.
J Biol Chem ; 288(44): 31602-15, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24047900

ABSTRACT

Chagas disease, caused by the eukaryotic (protozoan) parasite Trypanosoma cruzi, is an alarming emerging global health problem with no clinical drugs available to treat the chronic stage. Azole inhibitors of sterol 14α-demethylase (CYP51) were proven effective against Chagas, and antifungal drugs posaconazole and ravuconazole have entered clinical trials in Spain, Bolivia, and Argentina. Here we present the x-ray structures of T. cruzi CYP51 in complexes with two alternative drug candidates, pyridine derivatives (S)-(4-chlorophenyl)-1-(4-(4-(trifluoromethyl)phenyl)-piperazin-1-yl)-2-(pyridin-3-yl)ethanone (UDO; Protein Data Bank code 3ZG2) and N-[4-(trifluoromethyl)phenyl]-N-[1-[5-(trifluoromethyl)-2-pyridyl]-4-piperi-dyl]pyridin-3-amine (UDD; Protein Data Bank code 3ZG3). These compounds have been developed by the Drugs for Neglected Diseases initiative (DNDi) and are highly promising antichagasic agents in both cellular and in vivo experiments. The binding parameters and inhibitory effects on sterol 14α-demethylase activity in reconstituted enzyme reactions confirmed UDO and UDD as potent and selective T. cruzi CYP51 inhibitors. Comparative analysis of the pyridine- and azole-bound CYP51 structures uncovered the features that make UDO and UDD T. cruzi CYP51-specific. The structures suggest that although a precise fit between the shape of the inhibitor molecules and T. cruzi CYP51 active site topology underlies their high inhibitory potency, a longer coordination bond between the catalytic heme iron and the pyridine nitrogen implies a weaker influence of pyridines on the iron reduction potential, which may be the basis for the observed selectivity of these compounds toward the target enzyme versus other cytochrome P450s, including human drug-metabolizing P450s. These findings may pave the way for the development of novel CYP51-targeted drugs with optimized metabolic properties that are very much needed for the treatment of human infections caused by eukaryotic microbial pathogens.


Subject(s)
14-alpha Demethylase Inhibitors/chemistry , Antiprotozoal Agents/chemistry , Chagas Disease/enzymology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Sterol 14-Demethylase/chemistry , Trypanosoma cruzi/enzymology , 14-alpha Demethylase Inhibitors/therapeutic use , Antiprotozoal Agents/therapeutic use , Chagas Disease/drug therapy , Chagas Disease/genetics , Crystallography, X-Ray , Humans , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sterol 14-Demethylase/genetics , Sterol 14-Demethylase/metabolism , Thiazoles/chemistry , Triazoles/chemistry , Trypanosoma cruzi/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...