Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 673: 108080, 2019 09 30.
Article in English | MEDLINE | ID: mdl-31445894

ABSTRACT

The electron configuration of flavin cofactors, FMN and FAD, is a critical factor governing the reactivity of NADPH-cytochrome P450 reductase (CPR). The current view of electron transfer by the mammalian CPR, based on equilibrium redox potentials of the flavin cofactors, is that the two electron-reduced FMN hydroquinone (FMNH2), rather than one electron-reduced FMN semiquinone, serves as electron donor to the terminal protein acceptors. However, kinetic and thermodynamic studies on the CPR species originated from different organisms have shown that redox potentials measured at distinct electron transfer steps differ from redox potentials determined by equilibrium titration. Collectively, previous observations suggest that the short-lived transient semiquinone species may carry electrons in diflavin reductases. In this work, we have investigated spectroscopic properties of the CPR-bound FAD and FMN reduced at 77 K by radiolytically-generated thermalized electrons. Using UV-vis spectroscopy, we demonstrated that upon cryo-reduction of oxidized yeast CPR (yCPR) containing an equimolar ratio of both FAD and FMN, or FAD alone, neutral semiquinones were trapped at 77 K. During annealing at the elevated temperatures, unstable short-lived neutral semiquinones relaxed to spectroscopically distinct air-stable neutral semiquinones. This transition was independent of pH within the 6.0-10.7 range. Our data on yeast CPR are in line with the previous observations of others that the flavin short-lived transient semiquinone intermediates may have a role in the electron transfer by CPR at physiological conditions.


Subject(s)
Flavin-Adenine Dinucleotide/analogs & derivatives , NADPH-Ferrihemoprotein Reductase/metabolism , Temperature , Yeasts/enzymology , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Glucose Oxidase/metabolism , Kinetics , Oxidation-Reduction
2.
J Med Chem ; 62(3): 1330-1347, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30615444

ABSTRACT

We have designed and synthesized a series of new imidazole-based compounds structurally related to an antiprotozoal agent with nanomolar activity which we identified recently. The new analogues possess micromolar activities against Trypanosoma brucei rhodesiense and Leishmania donovani and nanomolar potency against Plasmodium falciparum. Most of the analogues displayed IC50 within the low nanomolar range against Trypanosoma cruzi, with very high selectivity toward the parasite. Discussion of structure-activity relationships and in vitro biological data for the new compounds are provided against a number of different protozoa. The mechanism of action for the most potent derivatives (5i, 6a-c, and 8b) was assessed by a target-based assay using recombinant T. cruzi CYP51. Bioavailability and efficacy of selected hits were assessed in a T. cruzi mouse model, where 6a and 6b reduced parasitemia in animals >99% following intraperitoneal administration of 25 mg/kg/day dose for 4 consecutive days.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Drug Design , Drug Evaluation, Preclinical , Imidazoles/chemistry , Imidazoles/pharmacology , Trypanosoma/drug effects , Animals , Antiprotozoal Agents/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Humans , Imidazoles/chemical synthesis , Parasitic Sensitivity Tests
3.
PLoS Pathog ; 14(9): e1007245, 2018 09.
Article in English | MEDLINE | ID: mdl-30212566

ABSTRACT

Naegleria fowleri is a free-living amoeba that can also act as an opportunistic pathogen causing severe brain infection, primary amebic meningoencephalitis (PAM), in humans. The high mortality rate of PAM (exceeding 97%) is attributed to (i) delayed diagnosis, (ii) lack of safe and effective anti-N. fowleri drugs, and (iii) difficulty of delivering drugs to the brain. Our work addresses identification of new molecular targets that may link anti-Naegleria drug discovery to the existing pharmacopeia of brain-penetrant drugs. Using inhibitors with known mechanism of action as molecular probes, we mapped the sterol biosynthesis pathway of N. fowleri by GC-MS analysis of metabolites. Based on this analysis, we chemically validated two enzymes downstream to CYP51, sterol C24-methyltransferase (SMT, ERG6) and sterol Δ8-Δ7 -isomerase (ERG2), as potential therapeutic drug targets in N. fowleri. The sterol biosynthetic cascade in N. fowleri displayed a mixture of canonical features peculiar to different domains of life: lower eukaryotes, plants and vertebrates. In addition to the cycloartenol→ergosterol biosynthetic route, a route leading to de novo cholesterol biosynthesis emerged. Isotopic labeling of the de novo-synthesized sterols by feeding N. gruberi trophozoites on the U13C-glucose-containing growth medium identified an exogenous origin of cholesterol, while 7-dehydrocholesterol (7DHC) had enriched 13C-content, suggesting a dual origin of this metabolite both from de novo biosynthesis and metabolism of scavenged cholesterol. Sterol homeostasis in Naegleria may be orchestrated over the course of its life-cycle by a "switch" between ergosterol and cholesterol biosynthesis. By demonstrating the growth inhibition and synergistic effects of the sterol biosynthesis inhibitors, we validated new, potentially druggable, molecular targets in N. fowleri. The similarity of the Naegleria sterol Δ8-Δ7 -isomerase to the human non-opioid σ1 receptor, implicated in human CNS conditions such as addiction, amnesia, pain and depression, provides an incentive to assess structurally diverse small-molecule brain-penetrant drugs targeting the human receptor for anti-Naegleria activity.


Subject(s)
Naegleria fowleri/metabolism , Sterols/biosynthesis , Amino Acid Sequence , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/pharmacology , Biosynthetic Pathways/drug effects , Blood-Brain Barrier , Central Nervous System Protozoal Infections/drug therapy , Central Nervous System Protozoal Infections/parasitology , Cholesterol/biosynthesis , Drug Discovery , Drug Repositioning , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Gas Chromatography-Mass Spectrometry , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Methyltransferases/metabolism , Naegleria fowleri/drug effects , Naegleria fowleri/pathogenicity , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sequence Homology, Amino Acid , Steroid Isomerases/antagonists & inhibitors , Steroid Isomerases/genetics , Steroid Isomerases/metabolism
4.
Eur J Med Chem ; 156: 53-60, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-30006174

ABSTRACT

We discovered a series of azole antifungal compounds as effective antiprotozoal agents. They displayed promising inhibitory activities within the micromolar-submicromolar range against P. falciparum, L. donovani, and T. b. rhodesiense. Moreover, most of such compounds showed excellent nanomolar IC50 against T. cruzi, showing also very low cytotoxicity. Discussion of structure-activity relationships and biological data for these compounds are provided against the different parasites. To assess the mechanism of action against T. cruzi we proved that the most potent compounds (3b, 3j-l) inhibited the T. cruzi CYP51. Moreover, the most active derivative 3j dramatically reduced parasitemia in T. cruzi mouse model without acute toxicity.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Chagas Disease/drug therapy , Imidazoles/chemistry , Imidazoles/pharmacology , Trypanosoma cruzi/drug effects , Animals , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/therapeutic use , Cell Line , Female , Humans , Imidazoles/chemical synthesis , Imidazoles/therapeutic use , Inhibitory Concentration 50 , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Malaria, Falciparum/drug therapy , Mice , Mice, Inbred BALB C , Parasitic Sensitivity Tests , Plasmodium falciparum/drug effects , Rats , Structure-Activity Relationship , Trypanosoma brucei rhodesiense/drug effects , Trypanosomiasis, African/drug therapy
5.
J Biol Chem ; 293(29): 11433-11446, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29858244

ABSTRACT

Cytochrome P450 4B1 (4B1) functions in both xenobiotic and endobiotic metabolism. An ester linkage between Glu-310 in 4B1 and the 5-methyl group of heme facilitates preferential hydroxylation of terminal (ω) methyl groups of hydrocarbons (HCs) and fatty acids compared with ω-1 sites bearing weaker C-H bonds. This preference is retained albeit diminished 4-fold for the E310A mutant, but the reason for this is unclear. Here, a crystal structure of the E310A-octane complex disclosed that noncovalent interactions maintain heme deformation in the absence of the ester linkage. Consistent with the lower symmetry of the heme, resonance Raman (RR) spectroscopy revealed large enhancements of RR peaks for high-spin HC complexes of 4B1 and the E310A mutant relative to P450 3A4. Whereas these enhancements were diminished in RR spectra of a low-spin 4B1-N-hydroxy-N'-(4-butyl-2-methylphenyl)formamidine complex, a crystal structure indicated that this inhibitor does not alter heme ruffling. RR spectra of Fe2+-CO HC complexes revealed larger effects of HC length in E310A than in 4B1, suggesting that reduced rigidity probably underlies increased E310A-catalyzed (ω-1)-hydroxylation. Diminished effects of the HC on the position of the Fe-CO stretching mode in 4B1 suggested that the ester linkage limits substrate access to the CO. Heme ruffling probably facilitates autocatalytic ester formation by reducing inhibitory coordination of Glu-310 with the heme iron. This also positions the 5-methyl for a reaction with the proposed glutamyl radical intermediate and potentially enhances oxo-ferryl intermediate reactivity for generation of the glutamyl radical to initiate ester bond formation and ω-hydroxylation.


Subject(s)
Aryl Hydrocarbon Hydroxylases/chemistry , Heme/chemistry , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Catalytic Domain , Crystallography, X-Ray , Heme/metabolism , Hydroxylation , Models, Molecular , Oxidation-Reduction , Rabbits , Spectrum Analysis, Raman , Stereoisomerism , Substrate Specificity
6.
J Inorg Biochem ; 184: 79-87, 2018 07.
Article in English | MEDLINE | ID: mdl-29684698

ABSTRACT

Cytochrome P450 19 (CYP19, aromatase) catalyzes the conversion of androgens to estrogens in a sequence of three reactions that each depend on NADPH and O2. Aromatase is a phylogenetically-ancient enzyme and its breadth of expression in other species has highlighted distinct physiological functions. In songbirds, estrogen production is required for programming the neural circuits controlling song and in the determination of sex in fish and reptiles. This work describes the expression, purification, and biophysical characterization of Aptenodytes forsteri (Emperor penguin, af) aromatase. Using human cytochrome P450 reductase as a redox partner, afCYP19 displayed similar substrate turnover and LC/MS/MS confirmed that afCYP19 catalyzes the transformations through the intermediates 19-hydroxy- and 19-oxo-androstenedione. Androstenedione and anastrozole had the highest affinity for the enzyme and were followed closely by 19-hydroxyandrostenedione and testosterone. The affinity of 19-oxo-androstenedione for afCYP19 was ten-fold lower. The time-dependent changes in the Soret bands observed in stopped-flow mixing experiments of the steroidal ligands and the inhibitor anastrozole with afCYP19 were best described by a two-step binding mechanism. In summary, these studies describe the first biophysical characterization of an avian aromatase that displays strikingly similar enzyme kinetics and ligand binding properties to the human enzyme and could serve as a convenient model system for studies of the enigmatic transformation of androgens to estrogens.


Subject(s)
Aromatase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Anastrozole/metabolism , Androstenedione/analogs & derivatives , Androstenedione/metabolism , Spectrum Analysis, Raman , Testosterone/metabolism
7.
PLoS Negl Trop Dis ; 11(12): e0006104, 2017 12.
Article in English | MEDLINE | ID: mdl-29284029

ABSTRACT

Primary Amoebic Meningoencephalitis (PAM) is caused by Naegleria fowleri, a free-living amoeba that occasionally infects humans. While considered "rare" (but likely underreported) the high mortality rate and lack of established success in treatment makes PAM a particularly devastating infection. In the absence of economic inducements to invest in development of anti-PAM drugs by the pharmaceutical industry, anti-PAM drug discovery largely relies on drug 'repurposing'-a cost effective strategy to apply known drugs for treatment of rare or neglected diseases. Similar to fungi, N. fowleri has an essential requirement for ergosterol, a building block of plasma and cell membranes. Disruption of sterol biosynthesis by small-molecule inhibitors is a validated interventional strategy against fungal pathogens of medical and agricultural importance. The N. fowleri genome encodes the sterol 14-demethylase (CYP51) target sharing ~35% sequence identity to fungal orthologues. The similarity of targets raises the possibility of repurposing anti-mycotic drugs and optimization of their usage for the treatment of PAM. In this work, we (i) systematically assessed the impact of anti-fungal azole drugs, known as conazoles, on sterol biosynthesis and viability of cultured N. fowleri trophozotes, (ii) identified the endogenous CYP51 substrate by mass spectrometry analysis of N. fowleri lipids, and (iii) analyzed the interactions between the recombinant CYP51 target and conazoles by UV-vis spectroscopy and x-ray crystallography. Collectively, the target-based and parasite-based data obtained in these studies validated CYP51 as a potentially 'druggable' target in N. fowleri, and conazole drugs as the candidates for assessment in the animal model of PAM.


Subject(s)
14-alpha Demethylase Inhibitors/pharmacology , Amebicides/pharmacology , Central Nervous System Protozoal Infections/drug therapy , Drug Repositioning , Naegleria fowleri/drug effects , Nitriles/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Antifungal Agents/pharmacology , Cell Proliferation/drug effects , Central Nervous System Protozoal Infections/mortality , Central Nervous System Protozoal Infections/parasitology , Disease Models, Animal , Humans , Microscopy, Electron, Transmission , Naegleria fowleri/ultrastructure , Sterol 14-Demethylase/metabolism , Sterols/biosynthesis , Trophozoites/drug effects , Trophozoites/ultrastructure
8.
ACS Chem Biol ; 12(2): 422-434, 2017 02 17.
Article in English | MEDLINE | ID: mdl-27977118

ABSTRACT

Recent advances in cell-based, high-throughput phenotypic screening have identified new chemical compounds that are active against eukaryotic pathogens. A challenge to their future development lies in identifying these compounds' molecular targets and binding modes. In particular, subsequent structure-based chemical optimization and target-based screening require a detailed understanding of the binding event. Here, we use directed evolution and whole-genome sequencing of a drug-sensitive S. cerevisiae strain to identify the yeast ortholog of TcCyp51, lanosterol-14-alpha-demethylase (TcCyp51), as the target of MMV001239, a benzamide compound with activity against Trypanosoma cruzi, the etiological agent of Chagas disease. We show that parasites treated with MMV0001239 phenocopy parasites treated with another TcCyp51 inhibitor, posaconazole, accumulating both lanosterol and eburicol. Direct drug-protein binding of MMV0001239 was confirmed through spectrophotometric binding assays and X-ray crystallography, revealing a binding site shared with other antitrypanosomal compounds that target Cyp51. These studies provide a new probe chemotype for TcCyp51 inhibition.


Subject(s)
14-alpha Demethylase Inhibitors/therapeutic use , Chagas Disease/drug therapy , Directed Molecular Evolution , Trypanocidal Agents/therapeutic use , 14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , Amino Acid Sequence , Chagas Disease/parasitology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Discovery , Gas Chromatography-Mass Spectrometry , Molecular Docking Simulation , Plasmodium falciparum/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Spectrophotometry, Ultraviolet , Sterol 14-Demethylase/drug effects , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology
9.
Mol Pharmacol ; 90(1): 42-51, 2016 07.
Article in English | MEDLINE | ID: mdl-27126611

ABSTRACT

Cytochrome P450 3A4 (CYP3A4) is the dominant P450 enzyme involved in human drug metabolism, and its inhibition may result in adverse interactions or, conversely, favorably reduce the systemic elimination rates of poorly bioavailable drugs. Herein we describe a spectroscopic investigation of the interaction of CYP3A4 with N-methylritonavir, an analog of ritonavir, widely used as a pharmacoenhancer. In contrast to ritonavir, the binding affinity of N-methylritonavir for CYP3A4 is pH-dependent. At pH <7.4, the spectra are definitively type I, whereas at pH ≥7.4 the spectra have split Soret bands, including a red-shifted component characteristic of a P450-carbene complex. Variable-pH UV-visible spectroscopy binding studies with molecular fragments narrows the source of this pH dependence to its N-methylthiazolium fragment. The C2 proton of this group is acidic, and variable-pH resonance Raman spectroscopy tentatively assigns it a pKa of 7.4. Hence, this fragment of N-methylritonavir is expected to be readily deprotonated under physiologic conditions to yield a thiazol-2-ylidene, which is an N-heterocyclic carbene that has high-affinity for and is presumed to be subsequently captured by the heme iron. This mechanism is supported by time-dependent density functional theory with an active site model that accurately reproduces distinguishing features of the experimental UV-visible spectra of N-methylritonavir bound to CYP3A4. Finally, density functional theory calculations support that this novel interaction is as strong as the tightest-binding azaheterocycles found in P450 inhibitors and could offer new avenues for inhibitor development.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A/metabolism , Heterocyclic Compounds/pharmacology , Methane/analogs & derivatives , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Heterocyclic Compounds/chemistry , Humans , Hydrogen-Ion Concentration , Ligands , Methane/chemistry , Methane/pharmacology , Models, Molecular , Protons , Quantum Theory , Ritonavir/chemistry , Ritonavir/pharmacology , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman , Titrimetry
10.
J Biol Chem ; 290(2): 1186-96, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25425647

ABSTRACT

Aromatase (CYP19A1), the enzyme that converts androgens to estrogens, is of significant mechanistic and therapeutic interest. Crystal structures and computational studies of this enzyme shed light on the critical role of Asp(309) in substrate binding and catalysis. These studies predicted an elevated pK(a) for Asp(309) and proposed that protonation of this residue was required for function. In this study, UV-visible absorption, circular dichroism, resonance Raman spectroscopy, and enzyme kinetics were used to study the impact of pH on aromatase structure and androstenedione binding. Spectroscopic studies demonstrate that androstenedione binding is pH-dependent, whereas, in contrast, the D309N mutant retains its ability to bind to androstenedione across the entire pH range studied. Neither pH nor mutation perturbed the secondary structure or heme environment. The origin of the observed pH dependence was further narrowed to the protonation equilibria of Asp(309) with a parallel set of spectroscopic studies using exemestane and anastrozole. Because exemestane interacts with Asp(309) based on its co-crystal structure with the enzyme, its binding is pH-dependent. Aromatase binding to anastrozole is pH-independent, consistent with the hypothesis that this ligand exploits a distinct set of interactions in the active site. In summary, we assign the apparent pK(a) of 8.2 observed for androstenedione binding to the side chain of Asp(309). To our knowledge, this work represents the first experimental assignment of a pK(a) value to a residue in a cytochrome P450. This value is in agreement with theoretical calculations (7.7-8.1) despite the reliance of the computational methods on the conformational snapshots provided by crystal structures.


Subject(s)
Aromatase/chemistry , Aspartic Acid/chemistry , Mutation , Protein Conformation , Androgens/chemistry , Androgens/metabolism , Aromatase/metabolism , Aspartic Acid/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray , Estrogens/chemistry , Estrogens/metabolism , Humans , Oxidation-Reduction , Spectrum Analysis, Raman
11.
J Inorg Biochem ; 136: 81-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24793591

ABSTRACT

The acidic residues of the "acid-alcohol pair" in CYP51 enzymes are uniformly replaced with histidine. Herein, we adopt the Mycobacterium tuberculosis (mt) enzyme as a model system to investigate these residues' roles in finely tuning the heme conformation, iron spin state, and formation and decay of the oxyferrous enzyme. Properties of the mtCYP51 and the T260A, T260V, and H259A mutants were interrogated using UV-Vis and resonance Raman spectroscopies. Evidence supports that these mutations induce comprehensive changes in the heme environment. The heme iron spin states are differentially sensitive to the binding of the substrate, dihydrolanosterol (DHL). DHL and clotrimazole perturb the local environments of the heme vinyl and propionate substituents. Molecular dynamics (MD) simulations of the DHL-enzyme complexes support that the observed perturbations are attributable to changes in the DHL binding mode. Furthermore, the rates of the oxyferrous formation were measured using stopped-flow methods. These studies demonstrate that both HT mutations and DHL modulate the rates of oxyferrous formation. Paradoxically, the binding rate to the H259A mutant-DHL complex was approximately four-fold that of mtCYP51, a phenomenon that is predicted to result from the creation of an additional diffusion channel from loss of the H259-E173 ion pair in the mutant. Oxyferrous enzyme auto-oxidation rates were relatively constant, with the exception of the T260V-DHL complex. MD simulations lead us to speculate that this behavior may be attributed to the distortion of the heme macrocycle by the substrate.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Mycobacterium tuberculosis/enzymology , Oxygen/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Catalytic Domain , Cytochrome P-450 Enzyme System/genetics , Histidine/chemistry , Histidine/genetics , Kinetics , Lanosterol/analogs & derivatives , Lanosterol/chemistry , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Binding , Spectrum Analysis, Raman , Threonine/chemistry , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...