Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 9(2): 221-238, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36606559

ABSTRACT

Mycobacterium tuberculosis cytochrome bd quinol oxidase (cyt bd), the alternative terminal oxidase of the respiratory chain, has been identified as playing a key role during chronic infection and presents a putative target for the development of novel antitubercular agents. Here, we report confirmation of successful heterologous expression of M. tuberculosis cytochrome bd. The heterologous M. tuberculosis cytochrome bd expression system was used to identify a chemical series of inhibitors based on the 2-aryl-quinolone pharmacophore. Cytochrome bd inhibitors displayed modest efficacy in M. tuberculosis growth suppression assays together with a bacteriostatic phenotype in time-kill curve assays. Significantly, however, inhibitor combinations containing our front-runner cyt bd inhibitor CK-2-63 with either cyt bcc-aa3 inhibitors (e.g., Q203) and/or adenosine triphosphate (ATP) synthase inhibitors (e.g., bedaquiline) displayed enhanced efficacy with respect to the reduction of mycobacterium oxygen consumption, growth suppression, and in vitro sterilization kinetics. In vivo combinations of Q203 and CK-2-63 resulted in a modest lowering of lung burden compared to treatment with Q203 alone. The reduced efficacy in the in vivo experiments compared to in vitro experiments was shown to be a result of high plasma protein binding and a low unbound drug exposure at the target site. While further development is required to improve the tractability of cyt bd inhibitors for clinical evaluation, these data support the approach of using small-molecule inhibitors to target multiple components of the branched respiratory chain of M. tuberculosis as a combination strategy to improve therapeutic and pharmacokinetic/pharmacodynamic (PK/PD) indices related to efficacy.


Subject(s)
Antitubercular Agents , Mycobacterium tuberculosis , Quinolones , Antitubercular Agents/pharmacology , Cytochromes/antagonists & inhibitors , Electron Transport Complex IV/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Quinolones/pharmacology
2.
J Med Chem ; 60(9): 3703-3726, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28304162

ABSTRACT

A high-throughput screen (HTS) was undertaken against the respiratory chain dehydrogenase component, NADH:menaquinone oxidoreductase (Ndh) of Mycobacterium tuberculosis (Mtb). The 11000 compounds were selected for the HTS based on the known phenothiazine Ndh inhibitors, trifluoperazine and thioridazine. Combined HTS (11000 compounds) and in-house screening of a limited number of quinolones (50 compounds) identified ∼100 hits and four distinct chemotypes, the most promising of which contained the quinolone core. Subsequent Mtb screening of the complete in-house quinolone library (350 compounds) identified a further ∼90 hits across three quinolone subtemplates. Quinolones containing the amine-based side chain were selected as the pharmacophore for further modification, resulting in metabolically stable quinolones effective against multi drug resistant (MDR) Mtb. The lead compound, 42a (MTC420), displays acceptable antituberculosis activity (Mtb IC50 = 525 nM, Mtb Wayne IC50 = 76 nM, and MDR Mtb patient isolates IC50 = 140 nM) and favorable pharmacokinetic and toxicological profiles.


Subject(s)
Mycobacterium tuberculosis/drug effects , Quinolones/chemical synthesis , Quinolones/pharmacology , Animals , Caco-2 Cells , Carbon-13 Magnetic Resonance Spectroscopy , Drug Design , Electron Transport/drug effects , Hep G2 Cells , High-Throughput Screening Assays , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/metabolism , Proton Magnetic Resonance Spectroscopy , Quinolones/chemistry , Quinolones/pharmacokinetics , Rats , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Toxicity Tests
3.
J Antimicrob Chemother ; 68(4): 869-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23228936

ABSTRACT

OBJECTIVES: Phenothiazines have been shown to exhibit in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb) and multidrug-resistant Mtb. They are predicted to target the genetically validated respiratory chain component type II NADH:quinone oxidoreductase (Ndh). Using a set of compounds containing the phenothiazine pharmacophore, we have (i) investigated whether chemical validation data support the molecular target and (ii) evaluated pharmacophore tractability for further drug development. METHODS: Recombinant Mtb Ndh was generated and its functionality confirmed by steady-state kinetics. Pharmacodynamic profiling of the phenothiazines, including antitubercular efficacy in aerobic and O2-limited conditions, time-kill assays and isobole analyses against first-line antituberculars, was performed. Potential mitochondrial toxicity was assessed in a modified HepG2 cell-line assay and against bovine cytochrome bc1. RESULTS: Steady-state kinetic analyses revealed a substrate preference for coenzyme Q2 and an inability to utilize NADPH. A positive correlation between recombinant Ndh inhibition and kill of aerobically cultured Mtb was observed, whilst enhanced potency was demonstrated in a hypoxic model. Time-kill studies revealed the phenothiazines to be bactericidal whilst isobolograms exposed antagonism with isoniazid, indicative of intracellular NADH/NAD(+) couple perturbation. At therapeutic levels, phenothiazine-mediated toxicity was appreciable; however, specific mitochondrial targeting was excluded. CONCLUSIONS: Data generated support the hypothesis that Ndh is the molecular target of phenothiazines. The favourable pharmacodynamic properties of the phenothiazines are consistent with a target product profile that includes activity against dormant/persistent bacilli, rapid bactericidal activity and activity against drug-resistant Mtb by a previously unexploited mode of action. These properties warrant further medicinal chemistry to improve potency and safety.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Phenothiazines/pharmacology , Antitubercular Agents/chemistry , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Microbial Viability/drug effects , Mycobacterium tuberculosis/physiology , Phenothiazines/chemistry
4.
Proc Natl Acad Sci U S A ; 109(21): 8298-303, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22566611

ABSTRACT

There is an urgent need for new antimalarial drugs with novel mechanisms of action to deliver effective control and eradication programs. Parasite resistance to all existing antimalarial classes, including the artemisinins, has been reported during their clinical use. A failure to generate new antimalarials with novel mechanisms of action that circumvent the current resistance challenges will contribute to a resurgence in the disease which would represent a global health emergency. Here we present a unique generation of quinolone lead antimalarials with a dual mechanism of action against two respiratory enzymes, NADH:ubiquinone oxidoreductase (Plasmodium falciparum NDH2) and cytochrome bc(1). Inhibitor specificity for the two enzymes can be controlled subtly by manipulation of the privileged quinolone core at the 2 or 3 position. Inhibitors display potent (nanomolar) activity against both parasite enzymes and against multidrug-resistant P. falciparum parasites as evidenced by rapid and selective depolarization of the parasite mitochondrial membrane potential, leading to a disruption of pyrimidine metabolism and parasite death. Several analogs also display activity against liver-stage parasites (Plasmodium cynomolgi) as well as transmission-blocking properties. Lead optimized molecules also display potent oral antimalarial activity in the Plasmodium berghei mouse malaria model associated with favorable pharmacokinetic features that are aligned with a single-dose treatment. The ease and low cost of synthesis of these inhibitors fulfill the target product profile for the generation of a potent, safe, and inexpensive drug with the potential for eventual clinical deployment in the control and eradication of falciparum malaria.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Pyridines/pharmacology , Quinolones/pharmacology , Animals , Antimalarials/chemistry , Cells, Cultured , Electron Transport/drug effects , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex III/antagonists & inhibitors , Hepatocytes/cytology , Hepatocytes/parasitology , Macaca mulatta , Malaria, Falciparum/parasitology , Male , Mice , Mice, Inbred Strains , Mitochondria/drug effects , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Plasmodium cynomolgi/drug effects , Plasmodium cynomolgi/growth & development , Plasmodium falciparum/growth & development , Pyridines/chemistry , Quinolones/chemistry
5.
J Med Chem ; 55(7): 3144-54, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22380711

ABSTRACT

Malaria is responsible for approximately 1 million deaths annually; thus, continued efforts to discover new antimalarials are required. A HTS screen was established to identify novel inhibitors of the parasite's mitochondrial enzyme NADH:quinone oxidoreductase (PfNDH2). On the basis of only one known inhibitor of this enzyme, the challenge was to discover novel inhibitors of PfNDH2 with diverse chemical scaffolds. To this end, using a range of ligand-based chemoinformatics methods, ~17000 compounds were selected from a commercial library of ~750000 compounds. Forty-eight compounds were identified with PfNDH2 enzyme inhibition IC(50) values ranging from 100 nM to 40 µM and also displayed exciting whole cell antimalarial activity. These novel inhibitors were identified through sampling 16% of the available chemical space, while only screening 2% of the library. This study confirms the added value of using multiple ligand-based chemoinformatic approaches and has successfully identified novel distinct chemotypes primed for development as new agents against malaria.


Subject(s)
Antimalarials/chemistry , Databases, Factual , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Quinone Reductases/antagonists & inhibitors , Antimalarials/pharmacology , Bayes Theorem , High-Throughput Screening Assays , Informatics , Parasitic Sensitivity Tests , Plasmodium falciparum/drug effects , Principal Component Analysis , Protozoan Proteins/chemistry , Quinone Reductases/chemistry
6.
J Med Chem ; 55(5): 1831-43, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22364416

ABSTRACT

A program was undertaken to identify hit compounds against NADH:ubiquinone oxidoreductase (PfNDH2), a dehydrogenase of the mitochondrial electron transport chain of the malaria parasite Plasmodium falciparum. PfNDH2 has only one known inhibitor, hydroxy-2-dodecyl-4-(1H)-quinolone (HDQ), and this was used along with a range of chemoinformatics methods in the rational selection of 17 000 compounds for high-throughput screening. Twelve distinct chemotypes were identified and briefly examined leading to the selection of the quinolone core as the key target for structure-activity relationship (SAR) development. Extensive structural exploration led to the selection of 2-bisaryl 3-methyl quinolones as a series for further biological evaluation. The lead compound within this series 7-chloro-3-methyl-2-(4-(4-(trifluoromethoxy)benzyl)phenyl)quinolin-4(1H)-one (CK-2-68) has antimalarial activity against the 3D7 strain of P. falciparum of 36 nM, is selective for PfNDH2 over other respiratory enzymes (inhibitory IC(50) against PfNDH2 of 16 nM), and demonstrates low cytotoxicity and high metabolic stability in the presence of human liver microsomes. This lead compound and its phosphate pro-drug have potent in vivo antimalarial activity after oral administration, consistent with the target product profile of a drug for the treatment of uncomplicated malaria. Other quinolones presented (e.g., 6d, 6f, 14e) have the capacity to inhibit both PfNDH2 and P. falciparum cytochrome bc(1), and studies to determine the potential advantage of this dual-targeting effect are in progress.


Subject(s)
Antimalarials/chemical synthesis , Plasmodium falciparum/enzymology , Quinolones/chemical synthesis , Quinone Reductases/antagonists & inhibitors , Administration, Oral , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Crystallography, X-Ray , Drug Design , Electron Transport Complex III/antagonists & inhibitors , Humans , In Vitro Techniques , Malaria/drug therapy , Male , Mice , Microsomes, Liver/metabolism , Models, Molecular , Parasitic Sensitivity Tests , Plasmodium berghei , Plasmodium falciparum/drug effects , Quinolones/chemistry , Quinolones/pharmacology , Structure-Activity Relationship
7.
J Med Chem ; 55(5): 1844-57, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22364417

ABSTRACT

Following a program undertaken to identify hit compounds against NADH:ubiquinone oxidoreductase (PfNDH2), a novel enzyme target within the malaria parasite Plasmodium falciparum, hit to lead optimization led to identification of CK-2-68, a molecule suitable for further development. In order to reduce ClogP and improve solubility of CK-2-68 incorporation of a variety of heterocycles, within the side chain of the quinolone core, was carried out, and this approach led to a lead compound SL-2-25 (8b). 8b has IC(50)s in the nanomolar range versus both the enzyme and whole cell P. falciparum (IC(50) = 15 nM PfNDH2; IC(50) = 54 nM (3D7 strain of P. falciparum) with notable oral activity of ED(50)/ED(90) of 1.87/4.72 mg/kg versus Plasmodium berghei (NS Strain) in a murine model of malaria when formulated as a phosphate salt. Analogues in this series also demonstrate nanomolar activity against the bc(1) complex of P. falciparum providing the potential added benefit of a dual mechanism of action. The potent oral activity of 2-pyridyl quinolones underlines the potential of this template for further lead optimization studies.


Subject(s)
Antimalarials/chemical synthesis , Plasmodium falciparum/enzymology , Pyridines/chemical synthesis , Quinolones/chemical synthesis , Quinone Reductases/antagonists & inhibitors , Administration, Oral , Animals , Antimalarials/chemistry , Antimalarials/pharmacology , Atovaquone/pharmacology , Crystallography, X-Ray , Cytochromes b/genetics , Drug Design , Drug Resistance , Humans , Malaria/drug therapy , Male , Mice , Microsomes, Liver/metabolism , Models, Molecular , Parasitic Sensitivity Tests , Plasmodium berghei , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Pyridines/chemistry , Pyridines/pharmacology , Quinolones/chemistry , Quinolones/pharmacology , Rats , Structure-Activity Relationship
8.
FEBS J ; 279(9): 1675-93, 2012 May.
Article in English | MEDLINE | ID: mdl-22356105

ABSTRACT

The novel cytochrome P450/redox partner fusion enzyme CYP116B1 from Cupriavidus metallidurans was expressed in and purified from Escherichia coli. Isolated CYP116B1 exhibited a characteristic Fe(II)CO complex with Soret maximum at 449 nm. EPR and resonance Raman analyses indicated low-spin, cysteinate-coordinated ferric haem iron at both 10 K and ambient temperature, respectively, for oxidized CYP116B1. The EPR of reduced CYP116B1 demonstrated stoichiometric binding of a 2Fe-2S cluster in the reductase domain. FMN binding in the reductase domain was confirmed by flavin fluorescence studies. Steady-state reduction of cytochrome c and ferricyanide were supported by both NADPH/NADH, with NADPH used more efficiently (K(m[NADPH]) = 0.9 ± 0.5 µM and K(m[NADH]) = 399.1 ± 52.1 µM). Stopped-flow studies of NAD(P)H-dependent electron transfer to the reductase confirmed the preference for NADPH. The reduction potential of the P450 haem iron was -301 ± 7 mV, with retention of haem thiolate ligation in the ferrous enzyme. Redox potentials for the 2Fe-2S and FMN cofactors were more positive than that of the haem iron. Multi-angle laser light scattering demonstrated CYP116B1 to be monomeric. Type I (substrate-like) binding of selected unsaturated fatty acids (myristoleic, palmitoleic and arachidonic acids) was shown, but these substrates were not oxidized by CYP116B1. However, CYP116B1 catalysed hydroxylation (on propyl chains) of the herbicides S-ethyl dipropylthiocarbamate (EPTC) and S-propyl dipropylthiocarbamate (vernolate), and the subsequent N-dealkylation of vernolate. CYP116B1 thus has similar thiocarbamate-oxidizing catalytic properties to Rhodoccocus erythropolis CYP116A1, a P450 involved in the oxidative degradation of EPTC.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Oxidoreductases/chemistry , Bacterial Proteins/chemistry , Cloning, Molecular , Cupriavidus/enzymology , Cyanides/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Electron Spin Resonance Spectroscopy , Herbicides/metabolism , Imidazoles/pharmacology , Iron-Sulfur Proteins/chemistry , Lasers , NADP/metabolism , Nitric Oxide/pharmacology , Recombinant Fusion Proteins/metabolism , Rhodococcus/enzymology , Scattering, Radiation , Spectrophotometry, Ultraviolet , Thermodynamics , Thiocarbamates/metabolism
9.
J Biol Chem ; 287(13): 9731-9741, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22282497

ABSTRACT

Atovaquone is an anti-malarial drug used in combination with proguanil (e.g. Malarone(TM)) for the curative and prophylactic treatment of malaria. Atovaquone, a 2-hydroxynaphthoquinone, is a competitive inhibitor of the quinol oxidation (Q(o)) site of the mitochondrial cytochrome bc(1) complex. Inhibition of this enzyme results in the collapse of the mitochondrial membrane potential, disruption of pyrimidine biosynthesis, and subsequent parasite death. Resistance to atovaquone in the field is associated with point mutations in the Q(o) pocket of cytochrome b, most notably near the conserved Pro(260)-Glu(261)-Trp(262)-Tyr(263) (PEWY) region in the ef loop). The effect of this mutation has been extensively studied in model organisms but hitherto not in the parasite itself. Here, we have performed a molecular and biochemical characterization of an atovaquone-resistant field isolate, TM902CB. Molecular analysis of this strain reveals the presence of the Y268S mutation in cytochrome b. The Y268S mutation is shown to confer a 270-fold shift of the inhibitory constant (K(i)) for atovaquone with a concomitant reduction in the V(max) of the bc(1) complex of ∼40% and a 3-fold increase in the observed K(m) for decylubiquinol. Western blotting analyses reveal a reduced iron-sulfur protein content in Y268S bc(1) suggestive of a weakened interaction between this subunit and cytochrome b. Gene expression analysis of the TM902CB strain reveals higher levels of expression, compared with the 3D7 (atovaquone-sensitive) control strain in bc(1) and cytochrome c oxidase genes. It is hypothesized that the observed differential expression of these and other key genes offsets the fitness cost resulting from reduced bc(1) activity.


Subject(s)
Antimalarials/pharmacology , Atovaquone/pharmacology , Cytochromes b/biosynthesis , Drug Resistance , Gene Expression Regulation, Enzymologic , Mutation, Missense , Plasmodium falciparum/enzymology , Protozoan Proteins/biosynthesis , Amino Acid Substitution , Cytochromes b/genetics , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Plasmodium falciparum/genetics , Proguanil/pharmacology , Protozoan Proteins/genetics
10.
J Med Chem ; 53(3): 1211-21, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20067272

ABSTRACT

Twenty-six novel naphthoquinone aliphatic esters were synthesized by esterification of 1,4-naphthoquinone alcohols with various aliphatic acids. The 1,4-naphthoquinone alcohols were prepared from 1-hydroxy-2-naphthoic acid in nine steps with excellent yields. Twenty-four of the novel synthetic naphthoquinone esters showed significant antimalarial activity with IC(50) values in the range of 0.03-16.63 microM. The length of the aliphatic chain and the presence of C-2' substituents on the propyl chain affected the activity. Interestingly, compounds 31 and 37 showed very good antimalarial activity and were not toxic to normal Vero cells, and the PTI values of 31 (>1990.38) and 37 (1825.94) are excellent. Both 31 and 37 showed potent inhibition against P. falciparum 3D7 cyt bc(1) and no inhibition on rat cyt bc(1). They showed IC(50) values in the nanomolar range, providing full inhibition of cyt bc(1) with one molecule inhibitor bound per cyt bc(1) monomer at the Q(o) site.


Subject(s)
Antimalarials/pharmacology , Antineoplastic Agents/pharmacology , Malaria, Falciparum/drug therapy , Naphthoquinones/chemistry , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Chlorocebus aethiops , Electron Transport Complex III/antagonists & inhibitors , Erythrocytes/drug effects , Inhibitory Concentration 50 , Malaria, Falciparum/parasitology , Male , Mitochondrial Membranes/drug effects , Rats , Rats, Wistar , Saccharomyces cerevisiae/growth & development , Structure-Activity Relationship , Vero Cells
11.
Methods Enzymol ; 456: 303-20, 2009.
Article in English | MEDLINE | ID: mdl-19348896

ABSTRACT

Type II NADH: quinone oxidoreductases (ndh) are flavoenzymes found in a broad range of organisms including plants, fungi, protozoa, and bacteria. The ndh enzymes catalyze the oxidation of NADH with concomitant reduction of quinone (Q). These membrane-bound respiratory enzymes differ from the canonical NADH: dehydrogenase (complex I), because they are not involved in the vectorial transfer of protons across membranes. In Plasmodium falciparum and Mycobacterium tuberculosis, causative agents of malaria and tuberculosis, respectively, ndhs have aroused interest because of the essential role played in maintaining a reduced Q-pool during infection. In this chapter, we present methods for the measurement of steady-state parameters for ndhs from both pathogens, highlighting best practices and caveats. In addition, owing to the interest in ndhs as potential chemotherapeutic targets, we describe a miniaturized endpoint assay that is validated for high-throughput screening (HTS) of chemical libraries.


Subject(s)
Mycobacterium tuberculosis/enzymology , Plasmodium falciparum/enzymology , Quinone Reductases/metabolism , Animals , Kinetics
12.
Biochemistry ; 45(27): 8427-43, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16819841

ABSTRACT

Mycobacterium tuberculosis encodes a P450 of the sterol demethylase family (CYP51) chromosomally located adjacent to a ferredoxin (Fdx). CYP51 and Fdx were purified to homogeneity and characterized. Spectroscopic analyses were consistent with cysteinate- and aqua-ligated heme iron in CYP51. An epsilon419 of 134 mM(-1) cm(-1) was determined for oxidized CYP51. Analysis of interactions of 1-, 2-, and 4-phenylimidazoles with CYP51 showed that the 1- and 4-forms were heme iron-coordinating inhibitors, while 2-phenylimidazole induced a substrate-like optical shift. The 2-phenyimidazole-bound CYP51 demonstrated unusual decreases in high-spin heme iron content at elevated temperatures and an almost complete absence of high-spin heme iron by low-temperature EPR. These data suggest thermally induced alterations in CYP51 active site structure and/or binding modes for the small ligand. Reduction of CYP51 in the presence of carbon monoxide leads to formation of an Fe(II)-CO complex with a Soret absorption maximum at 448.5 nm, which collapses (at 0.246 min(-1) at pH 7.0) forming a species with a Soret maximum at 421.5 nm (the inactive P420 form). The rate of P420 formation is accelerated at lower pH, consistent with protonation of the cysteinate (Cys 394) to a thiol underlying the P450-P420 transition. The P450 form is stabilized by estriol, which induces a type I spectral shift on binding CYP51 (Kd = 21.7 microM). Nonstandard spectral changes occur on CYP51 reduction (using either dithionite or natural redox partners), including a blue-shifted Soret band and development of a strong feature at approximately 558.5 nm, suggestive of cysteine thiol ligation. Thus, ligand-free ferrous CYP51 is prone to thiolate ligand protonation even in the absence of carbon monoxide. Analysis of reoxidized CYP51 demonstrates that the enzyme re-forms P450, indicating that Cys 394 thiol is readily deprotonated to thiolate in the ferric form. Spectroscopic analysis of Fdx by EPR (resonance at g = 2.03) and magnetic CD (intensity for oxidized and reduced forms and signal intensity dependence on field strength and temperature) demonstrated that Fdx binds a [3Fe-4S] iron-sulfur cluster. Potentiometric studies show that the midpoint potential for ligand-free CYP51 is -375 mV, increasing to -225 mV in the estriol-bound form. The Fdx potential is -31 mV. Fdx forms a productive electron transfer complex with CYP51 and reduces it at a rate of 3.0 min(-1) in the ligand-free form and 4.3 min(-1) in the estriol-bound form, despite a thermodynamic barrier. Steady-state analysis of a M. tuberculosis class I redox system comprising flavoprotein reductase A (FprA), Fdx, and estriol-bound CYP51 indicates heme iron reduction as a rate-limiting step.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome P-450 Enzyme System/chemistry , Ferredoxins/chemistry , Mycobacterium tuberculosis/enzymology , Oxidoreductases/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Carbon Monoxide/chemistry , Cytochrome P-450 Enzyme System/genetics , Heme/chemistry , Hydrogen-Ion Concentration , Iron/chemistry , Kinetics , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases/genetics , Potentiometry , Spectrum Analysis , Sterol 14-Demethylase
SELECTION OF CITATIONS
SEARCH DETAIL
...