Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Vaccine ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38704256

ABSTRACT

Mucosal vaccines have the potential to elicit protective immune responses at the point of entry of respiratory pathogens, thus preventing even the initial seed infection. Unlike licensed injectable vaccines, mucosal vaccines comprising protein subunits are only in development. One of the primary challenges associated with mucosal vaccines has been identifying and characterizing safe yet effective mucosal adjuvants that can effectively prime multi-factorial mucosal immunity. In this study, we tested NanoSTING, a liposomal formulation of the endogenous activator of the stimulator of interferon genes (STING) pathway, cyclic guanosine adenosine monophosphate (cGAMP), as a mucosal adjuvant. We formulated a vaccine based on the H1 antigen (fusion protein of Ag85b and ESAT-6) adjuvanted with NanoSTING. Intranasal immunization of NanoSTING-H1 elicited a strong T-cell response in the lung of vaccinated animals characterized by (a) CXCR3+ KLRG1- lung resident T cells that are known to be essential for controlling bacterial infection, (b) IFNγ-secreting CD4+ T cells which is necessary for intracellular bactericidal activity, and (c) IL17-secreting CD4+ T cells that can confer protective immunity against multiple clinically relevant strains of Mtb. Upon challenge with aerosolized Mycobacterium tuberculosis Erdman strain, intranasal NanoSTING-H1 provides protection comparable to subcutaneous administration of the live attenuated Mycobacterium bovis vaccine strain Bacille-Calmette-Guérin (BCG). Our results indicate that NanoSTING adjuvanted protein vaccines can elicit a multi-factorial immune response that protects from infection by M. tuberculosis.

2.
J Biol Chem ; 295(17): 5785-5794, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32169899

ABSTRACT

The Mycobacterium tuberculosis virulence factor EsxA and its chaperone EsxB are secreted as a heterodimer (EsxA:B) and are crucial for mycobacterial escape from phagosomes and cytosolic translocation. Current findings support the idea that for EsxA to interact with host membranes, EsxA must dissociate from EsxB at low pH. However, the molecular mechanism by which the EsxA:B heterodimer separates is not clear. In the present study, using liposome-leakage and cytotoxicity assays, LC-MS/MS-based proteomics, and CCF-4 FRET analysis, we obtained evidence that the Nα-acetylation of the Thr-2 residue on EsxA, a post-translational modification that is present in mycobacteria but absent in Escherichia coli, is required for the EsxA:B separation. Substitutions at Thr-2 that precluded Nα-acetylation inhibited the heterodimer separation and hence prevented EsxA from interacting with the host membrane, resulting in attenuated mycobacterial cytosolic translocation and virulence. Molecular dynamics simulations revealed that at low pH, the Nα-acetylated Thr-2 makes direct and frequent "bind-and-release" contacts with EsxB, which generates a force that pulls EsxB away from EsxA. In summary, our findings provide evidence that the Nα-acetylation at Thr-2 of EsxA facilitates dissociation of the EsxA:B heterodimer required for EsxA membrane permeabilization and mycobacterial cytosolic translocation and virulence.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cytosol/metabolism , Mycobacterium tuberculosis/physiology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/metabolism , Acetylation , Animals , Antigens, Bacterial/analysis , Bacterial Proteins/analysis , Cell Membrane/metabolism , Host-Pathogen Interactions , Humans , Mice , Molecular Dynamics Simulation , Mycobacterium tuberculosis/chemistry , Protein Multimerization , RAW 264.7 Cells , Tuberculosis/microbiology , Virulence , Virulence Factors/analysis , Virulence Factors/metabolism
3.
Sci Rep ; 6: 32618, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27600772

ABSTRACT

EsxA is required for virulence of Mycobacterium tuberculosis (Mtb) and plays an essential role in phagosome rupture and translocation to the cytosol of macrophages. Recent biochemical studies have demonstrated that EsxA is a membrane-permeabilizing protein. However, evidence that link EsxA membrane-permeabilizing activity to Mtb cytosolic translocation and virulence is lacking. Here we found that mutations at glutamine 5 (Q5) could up or down regulate EsxA membrane-permeabilizing activity. The mutation Q5K significantly diminished the membrane-permeabilizing activity, while Q5V enhanced the activity. By taking advantage of the single-residue mutations, we tested the effects of EsxA membrane-permeabilizing activity on mycobacterial virulence and cytosolic translocation using the esxA/esxB knockout strains of Mycobacterium marinum (Mm) and Mtb. Compared to wild type (WT), the Q5K mutant exhibited significantly attenuated virulence, evidenced by intracellular survival and cytotoxicity in mouse macrophages as well as infection of zebra fish embryos. The attenuated virulence of the Q5K mutant was correlated to the impaired cytosolic translocation. On the contrary, the Q5V mutant had a significantly increased cytosolic translocation and showed an overall increased virulence. This study provides convincing evidence that EsxA contributes to mycobacterial virulence with its membrane-permeabilizing activity that is required for cytosolic translocation.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cytosol/metabolism , Glutamine/genetics , Mutation/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/metabolism , Bacterial Adhesion/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Line , Cell Membrane Permeability , Down-Regulation/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Homologous Recombination/genetics , Humans , Mice , Microbial Viability/genetics , Mycobacterium tuberculosis/growth & development , Protein Transport , Up-Regulation/genetics , Virulence , Zebrafish/embryology
4.
Biomed Opt Express ; 5(11): 3990-4001, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25426325

ABSTRACT

Transition from latency to active tuberculosis requires Mycobacterium tuberculosis (Mtb) to penetrate the phagosomal membrane and translocate to the cytosol of the host macrophage. Quantitative two-photon fluorescence resonance energy transfer (FRET) microscopy is developed to measure cytosolic translocation using Mycobacterium marinum (Mm) as a model organism for Mtb. Macrophages were infected with Mm or non-pathogenic Mycobacterium smegmatis (Ms) as a control, then loaded with a FRET substrate. Once translocation occurs, mycobacterium-bearing ß-lactamase cleaves the substrate, resulting in decrease of FRET signal. Quantification of this FRET signal change revealed that Mm, but not Ms, is capable of translocating to the cytosol.

5.
Methods Mol Biol ; 987: 79-94, 2013.
Article in English | MEDLINE | ID: mdl-23475669

ABSTRACT

Tuberculosis remains a leading cause of human mortality. The emergence of strains of Mycobacterium tuberculosis (Mtb), the causative agent, that are resistant to first- and second-line antitubercular drugs urges the development of new therapeutics. The genome of Mtb encodes 20 cytochrome P450 enzymes, at least some of which are potential candidates (CYP121, CYP125, and CYP128) for drug targeting. In this regard, we examined the specific role of CYP125 in the cholesterol degradation pathway, using genetic and mass spectrometric approaches. The analysis of lipid profiles from Mtb cells grown on cholesterol revealed that CYP125, by virtue of its C26-monooxygenase activity, is essential for cholesterol degradation, and, consequently, for the incorporation of side-chain carbon atoms into cellular lipids, as evidenced by an increase in the mass of the methyl-branched phthiocerol dimycocerosates (PDIM). Moreover, this work also led to the identification of cholest-4-en-3-one as a source of cellular toxicity. Herein, we describe the experimental procedures that led to elucidation of the physiological function of CYP125. A similar approach can be used to study other important Mtb P450 enzymes.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Mass Spectrometry/methods , Mycobacterium tuberculosis/enzymology , Blotting, Southern , Cholesterol/isolation & purification , Cholesterol/metabolism , Cloning, Molecular , Cytochrome P-450 Enzyme System/genetics , DNA, Bacterial/genetics , Genome, Bacterial/genetics , Mutation , Mycobacteriophages/physiology , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/virology , Transduction, Genetic , Virus Assembly
6.
J Am Chem Soc ; 134(15): 6673-84, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22444582

ABSTRACT

CYP125 from Mycobacterium tuberculosis catalyzes sequential oxidation of the cholesterol side-chain terminal methyl group to the alcohol, aldehyde, and finally acid. Here, we demonstrate that CYP125 simultaneously catalyzes the formation of five other products, all of which result from deformylation of the sterol side chain. The aldehyde intermediate is shown to be the precursor of both the conventional acid metabolite and the five deformylation products. The acid arises by protonation of the ferric-peroxo anion species and formation of the ferryl-oxene species, also known as Compound I, followed by hydrogen abstraction and oxygen transfer. The deformylation products arise by addition of the same ferric-peroxo anion to the aldehyde intermediate to give a peroxyhemiacetal that leads to C-C bond cleavage. This bifurcation of the catalytic sequence has allowed us to examine the effect of electron donation by the proximal ligand on the properties of the ferric-peroxo anion. Replacement of the cysteine thiolate iron ligand by a selenocysteine results in UV-vis, EPR, and resonance Raman spectral changes indicative of an increased electron donation from the proximal selenolate ligand to the iron. Analysis of the product distribution in the reaction of the selenocysteine substituted enzyme reveals a gain in the formation of the acid (Compound I pathway) at the expense of deformylation products. These observations are consistent with an increase in the pK(a) of the ferric-peroxo anion, which favors its protonation and, therefore, Compound I formation.


Subject(s)
Biocatalysis , Cytochrome P-450 Enzyme System/chemistry , Electrons , Ligands , Mycobacterium tuberculosis/enzymology
7.
Trends Microbiol ; 19(11): 530-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21924910

ABSTRACT

Mycobacterium tuberculosis (Mtb) is an intracellular pathogen that infects 10 million people worldwide and kills 2 million people every year. The uptake and utilization of nutrients by Mtb within the host cell is still poorly understood, although lipids play an important role in Mtb persistence. The recent identification of a large regulon of cholesterol catabolic genes suggests that Mtb can use host sterol for infection and persistence. In this review, we report on recent progress in elucidation of the Mtb cholesterol catabolic reactions and their potential utility as targets for tuberculosis therapeutic agents.


Subject(s)
Cholesterol/biosynthesis , Host-Pathogen Interactions , Mycobacterium tuberculosis/metabolism , Tuberculosis/metabolism , Animals , Anticholesteremic Agents/pharmacology , Biosynthetic Pathways/drug effects , Cholesterol/chemistry , Humans , Mycobacterium tuberculosis/genetics , Tuberculosis/drug therapy , Tuberculosis/genetics , Tuberculosis/microbiology
8.
Biochemistry ; 50(14): 3014-24, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21381758

ABSTRACT

Cytochrome P450-mediated monooxygenation generally proceeds via a reactive ferryl intermediate coupled to a ligand radical [Fe(IV)═O]+• termed Compound I (Cpd I). The proximal cysteine thiolate ligand is a critical determinant of the spectral and catalytic properties of P450 enzymes. To explore the effect of an increased level of donation of electrons by the proximal ligand in the P450 catalytic cycle, we recently reported successful incorporation of SeCys into the active site of CYP119, a thermophilic cytochrome P450. Here we report relevant physical properties of SeCYP119 and a detailed analysis of the reaction of SeCYP119 with m-chloroperbenzoic acid. Our results indicate that the selenolate anion reduces rather than stabilizes Cpd I and also protects the heme from oxidative destruction, leading to the generation of a new stable species with an absorbance maximum at 406 nm. This stable intermediate can be returned to the normal ferric state by reducing agents and thiols, in agreement with oxidative modification of the selenolate ligand itself. Thus, in the seleno protein, the oxidative damage shifts from the heme to the proximal ligand, presumably because (a) an increased level of donation of electrons more efficiently quenches reactive species such as Cpd I and (b) the protection of the thiolate ligand provided by the protein active site structure is insufficient to shield the more oxidizable selenolate ligand.


Subject(s)
Archaeal Proteins/chemistry , Chlorobenzoates/chemistry , Cytochrome P-450 Enzyme System/chemistry , Selenocysteine/chemistry , Archaeal Proteins/metabolism , Chlorobenzoates/metabolism , Cytochrome P-450 Enzyme System/metabolism , Ferric Compounds/chemistry , Ferrous Compounds/chemistry , Kinetics , Oxidation-Reduction , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry
9.
Arch Biochem Biophys ; 507(1): 86-94, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-20727847

ABSTRACT

The regiospecific or preferential ω-hydroxylation of hydrocarbon chains is thermodynamically disfavored because the ease of C-H bond hydroxylation depends on the bond strength, and the primary C-H bond of a terminal methyl group is stronger than the secondary or tertiary C-H bond adjacent to it. The hydroxylation reaction will therefore occur primarily at the adjacent secondary or tertiary C-H bond unless the protein structure specifically enforces primary C-H bond oxidation. Here we review the classes of enzymes that catalyze ω-hydroxylation and our current understanding of the structural features that promote the ω-hydroxylation of unbranched and methyl-branched hydrocarbon chains. The evidence indicates that steric constraints are used to favor reaction at the ω-site rather than at the more reactive (ω-1)-site.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fatty Acids/metabolism , Animals , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cytochrome P-450 Enzyme System/chemistry , Fatty Acids/chemistry , Humans , Hydroxylation , Models, Molecular , Molecular Structure , Substrate Specificity
10.
Bioorg Med Chem Lett ; 21(1): 332-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21109436

ABSTRACT

Cytochrome P450 CYP125A1 of Mycobacterium tuberculosis, a potential therapeutic target for tuberculosis in humans, initiates degradation of the aliphatic chain of host cholesterol and is essential for establishing M. tuberculosis infection in a mouse model of disease. We explored the interactions of CYP125A1 with a reverse type I inhibitor by X-ray structure analysis and UV-vis spectroscopy. Compound LP10 (α-[(4-methylcyclohexyl)carbonyl amino]-N-4-pyridinyl-1H-indole-3-propanamide), previously identified as a potent type II inhibitor of Trypanosomacruzi CYP51, shifts CYP125A1 to a water-coordinated low-spin state upon binding with low micromolar affinity. When LP10 is present in the active site, the crystal structure and spectral characteristics both demonstrate changes in lipophilic and electronic properties favoring coordination of the iron axial water ligand. These results provide an insight into the structural requirements for developing selective CYP125A1 inhibitors.


Subject(s)
Antitubercular Agents/chemistry , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Binding Sites , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Protein Binding , Spectrophotometry, Ultraviolet
11.
J Biol Chem ; 285(47): 36352-60, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20843794

ABSTRACT

One challenge to the development of new antitubercular drugs is the existence of multiple virulent strains that differ genetically. We and others have recently demonstrated that CYP125A1 is a steroid C(26)-monooxygenase that plays a key role in cholesterol catabolism in Mycobacterium tuberculosis CDC1551 but, unexpectedly, not in the M. tuberculosis H37Rv strain. This discrepancy suggests that the H37Rv strain possesses compensatory activities. Here, we examined the roles in cholesterol metabolism of two other cytochrome P450 enzymes, CYP124A1 and CYP142A1. In vitro analysis, including comparisons of the binding affinities and catalytic efficiencies, demonstrated that CYP142A1, but not CYP124A1, can support the growth of H37Rv cells on cholesterol in the absence of cyp125A1. All three enzymes can oxidize the sterol side chain to the carboxylic acid state by sequential oxidation to the alcohol, aldehyde, and acid. Interestingly, CYP125A1 generates oxidized sterols of the (25S)-26-hydroxy configuration, whereas the opposite 25R stereochemistry is obtained with CYP124A1 and CYP142A1. Western blot analysis indicated that CYP124A1 was not detectably expressed in either the H37Rv or CDC1551 strains, whereas CYP142A1 was found in H37Rv but not CDC1551. Genetic complementation of CDC1551 Δcyp125A1 cells with the cyp124A1 or cyp142A1 genes revealed that the latter can fully rescue the growth defect on cholesterol, whereas cells overexpressing CYP124A1 grow poorly and accumulate cholest-4-en-3-one. Our data clearly establish a functional redundancy in the essential C(26)-monooxygenase activity of M. tuberculosis and validate CYP125A1 and CYP142A1 as possible drug targets.


Subject(s)
Cholestenones/metabolism , Cholesterol/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/enzymology , Blotting, Western , Cholestenones/chemistry , Cholesterol/chemistry , Cytochrome P-450 Enzyme System/genetics , Genetic Complementation Test , Hydroxylation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Tuberculosis/genetics , Tuberculosis/metabolism
12.
Chem Biol ; 17(8): 841-51, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20797613

ABSTRACT

We present the X-ray structure of PimD, both substrate-free and in complex with 4,5-desepoxypimaricin. PimD is a cytochrome P450 monooxygenase with native epoxidase activity that is critical in the biosynthesis of the polyene macrolide antibiotic pimaricin. Intervention in this secondary metabolic pathway could advance the development of drugs with improved pharmacologic properties. Epoxidation by P450 typically includes formation of a charge-transfer complex between an oxoferryl pi-cation radical species (Compound I) and the olefin pi-bond as the initial intermediate. Catalytic and structural evidence presented here suggest that epoxidation of 4,5-desepoxypimaricin proceeds via a hydroperoxoferric intermediate (Compound 0). The oxygen atom of Compound 0 distal to the heme iron may insert into the double bond of the substrate to make an epoxide ring. Stereoelectronic features of the putative transition state suggest substrate-assisted proton delivery.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Epoxy Compounds/metabolism , Natamycin/chemistry , Natamycin/metabolism , Amino Acid Sequence , Benzene/chemistry , Benzene/metabolism , Biocatalysis , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Peroxynitrous Acid/metabolism , Protein Conformation , Protons , Stereoisomerism , Substrate Specificity
13.
Mol Microbiol ; 77(3): 730-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20545858

ABSTRACT

The infectivity and persistence of Mycobacterium tuberculosis requires the utilization of host cell cholesterol. We have examined the specific role of cytochrome P450 CYP125A1 in the cholesterol degradation pathway using genetic, biochemical and high-resolution mass spectrometric approaches. The analysis of lipid profiles from cells grown on cholesterol revealed that CYP125A1 is required to incorporate the cholesterol side-chain carbon atoms into cellular lipids, as evidenced by an increase in the mass of the methyl-branched phthiocerol dimycocerosates. We observed that cholesterol-exposed cells lacking CYP125A1 accumulate cholest-4-en-3-one, suggesting that this is a physiological substrate for this enzyme. Reconstitution of enzymatic activity with spinach ferredoxin and ferredoxin reductase revealed that recombinant CYP125A1 indeed binds both cholest-4-en-3-one and cholesterol, efficiently hydroxylates both of them at C-27, and then further oxidizes 27-hydroxycholest-4-en-3-one to cholest-4-en-3-one-27-oic acid. We determined the X-ray structure of cholest-4-en-3-one-bound CYP125A1 at a resolution of 1.58 A. CYP125A1 is essential for growth of CDC1551 in media containing cholesterol or cholest-4-en-3-one. In its absence, the latter compound is toxic for both CDC1551 and H37Rv when added with glycerol as a second carbon source. CYP125A1 is a key enzyme in cholesterol metabolism and plays a crucial role in circumventing the deleterious effect of cholest-4-en-3-one.


Subject(s)
Bacterial Proteins/metabolism , Cholestenones/metabolism , Mycobacterium tuberculosis/enzymology , Steroid Hydroxylases/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Molecular Conformation , Molecular Sequence Data , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Protein Binding , Steroid Hydroxylases/chemistry , Steroid Hydroxylases/genetics
14.
Arch Biochem Biophys ; 493(1): 82-95, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19635450

ABSTRACT

Tuberculosis remains a leading cause of human mortality. The emergence of strains of Mycobacterium tuberculosis, the causative agent, that are resistant to the major frontline antitubercular drugs increases the urgency for the development of new therapeutic agents. Sequencing of the M. tuberculosis genome revealed the existence of 20 cytochrome P450 enzymes, some of which are potential candidates for drug targeting. The recent burst of studies reporting microarray-based gene essentiality and transcriptome analyses under in vitro, ex vivo and in vivo conditions highlight the importance of selected P450 isoforms for M. tuberculosis viability and pathogenicity. Current knowledge of the structural and biochemical properties of the M. tuberculosis P450 enzymes and their putative redox partners is reviewed, with an emphasis on findings related to their physiological function(s) as well as their potential as drug targets.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/enzymology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Genomics , Models, Molecular , Oxidation-Reduction , Phylogeny , Protein Conformation , Proteomics
15.
J Biol Chem ; 284(37): 25211-9, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19605350

ABSTRACT

The Mycobacterium tuberculosis P450 enzymes are of interest for their pharmacological development potential, as evidenced by their susceptibility to inhibition by antifungal azole drugs that normally target sterol 14alpha-demethylase (CYP51). Although antifungal azoles show promise, direct screening of compounds against M. tuberculosis P450 enzymes may identify novel, more potent, and selective inhibitory scaffolds. Here we report that CYP130 from M. tuberculosis has a natural propensity to bind primary arylamines with particular chemical architectures. These compounds were identified via a high throughput screen of CYP130 with a library of synthetic organic molecules. As revealed by subsequent x-ray structure analysis, selected compounds bind in the active site by Fe-coordination and hydrogen bonding of the arylamine group to the carbonyl oxygen of Gly(243). As evidenced by the binding of structural analogs, the primary arylamine group is indispensable, but synergism due to hydrophobic contacts between the rest of the molecule and protein amino acid residues is responsible for a binding affinity comparable with that of the antifungal azole drugs. The topology of the CYP130 active site favors angular coordination of the arylamine group over the orthogonal coordination of azoles. Upon substitution of Gly(243) by an alanine, the binding mode of azoles and some arylamines reverted from type II to type I because of hydrophobic and steric interactions with the alanine side chain. We suggest a role for the conserved Ala(Gly)(243)-Gly(244) motif in the I-helix in modulating both the binding affinity of the axial water ligand and the ligand selectivity of cytochrome P450 enzymes.


Subject(s)
Amines/chemistry , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray/methods , Gene Library , Ligands , Molecular Conformation , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Spectrophotometry/methods
16.
Biochemistry ; 48(5): 863-72, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19146393

ABSTRACT

During the initial growth infection stage of Mycobacterium tuberculosis (Mtb), (*)NO produced by host macrophages inhibits heme-containing terminal cytochrome oxidases, inactivates iron/sulfur proteins, and promotes entry into latency. Here we evaluate the potential of (*)NO as an inhibitor of Mtb cytochrome P450 enzymes, as represented by CYP130, CYP51, and the two previously uncharacterized enzymes CYP125 and CYP142. Using UV-visible absorption, resonance Raman, and stopped-flow spectroscopy, we investigated the reactions of (*)NO with these heme proteins in their ferric resting form. (*)NO coordinates tightly to CYP125 and CYP142 (submicromolar) and with a lower affinity (micromolar) to CYP130 and CYP51. Anaerobic reduction of the ferric-NO species with sodium dithionite led to the formation of two spectrally distinct classes of five-coordinate ferrous-NO complexes. Exposure of these species to O(2) revealed that the ferrous-NO forms of CYP125 and CYP142 are labile and convert back to the ferric state within a few minutes, whereas ferrous CYP130 and CYP51 bind (*)NO almost irreversibly. This work clearly indicates that, at physiological concentrations (approximately 1 microM), (*)NO would impair the activity of CYP130 and CYP51, whereas CYP125 and CYP142 are more resistant. Selective P450 inhibition may contribute to the inhibitory effects of (*)NO on Mtb growth.


Subject(s)
Bacterial Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/enzymology , Nitric Oxide/metabolism , Animals , Cattle , Ferric Compounds/metabolism , Horses , Mycobacterium tuberculosis/growth & development , Nitric Oxide/biosynthesis , Protein Binding , Reactive Nitrogen Species/biosynthesis , Reactive Nitrogen Species/metabolism , Recombinant Proteins/metabolism
17.
J Inorg Biochem ; 103(3): 316-25, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135260

ABSTRACT

To characterize heme oxygenase with a selenocysteine (SeCys) as the proximal iron ligand, we have expressed truncated human heme oxygenase-1 (hHO-1) His25Cys, in which Cys-25 is the only cysteine, in the Escherichia coli cysteine auxotroph strain BL21(DE3)cys. Selenocysteine incorporation into the protein was demonstrated by both intact protein mass measurement and mass spectrometric identification of the selenocysteine-containing tryptic peptide. One selenocysteine was incorporated into approximately 95% of the expressed protein. Formation of an adduct with Ellman's reagent (DTNB) indicated that the selenocysteine in the expressed protein was in the reduced state. The heme-His25SeCys hHO-1 complex could be prepared by either (a) supplementing the overexpression medium with heme, or (b) reconstituting the purified apoprotein with heme. Under reducing conditions in the presence of imidazole, a covalent bond is formed by addition of the selenocysteine residue to one of the heme vinyl groups. No covalent bond is formed when the heme is replaced by mesoheme, in which the vinyls are replaced by ethyl groups. These results, together with our earlier demonstration that external selenolate ligands can transfer an electron to the iron [Y. Jiang, P.R. Ortiz de Montellano, Inorg. Chem. 47 (2008) 3480-3482 ], indicate that a selenyl radical is formed in the hHO-1 His25SeCys mutant that adds to a heme vinyl group.


Subject(s)
Heme Oxygenase-1/metabolism , Heme/metabolism , Iron/metabolism , Selenocysteine/metabolism , Apoproteins/isolation & purification , Apoproteins/metabolism , Heme Oxygenase-1/genetics , Humans , Ligands , Plasmids/genetics , Plasmids/metabolism
18.
J Biol Chem ; 284(9): 5723-30, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19124459

ABSTRACT

The cytochrome P-450 PikC from Streptomyces venezuelae exhibits significant substrate tolerance and performs multiple hydroxylation reactions on structurally variant macrolides bearing the deoxyamino sugar desosamine. In previously determined co-crystal structures (Sherman, D. H., Li, S., Yermalitskaya, L. V., Kim, Y., Smith, J. A., Waterman, M. R., and Podust, L. M. (2006) J. Biol. Chem. 281, 26289-26297), the desosamine moiety of the native substrates YC-17 and narbomycin is bound in two distinct buried and surface-exposed binding pockets, mediated by specific interactions between the protonated dimethylamino group and the acidic amino acid residues Asp(50), Glu(85), and Glu(94). Although the Glu(85) and Glu(94) negative charges are essential for maximal catalytic activity of native enzyme, elimination of the surface-exposed negative charge at Asp(50) results in significantly enhanced catalytic activity. Nevertheless, the D50N substitution could not rescue catalytic activity of PikC(E94Q) based on lack of activity in the corresponding double mutant PikC(D50N/E94Q). To address the specific role for each desosamine-binding pocket, we analyzed the x-ray structures of the PikC(D50N) mutant co-crystallized with narbomycin (1.85A resolution) and YC-17 (3.2A resolution). In PikC(D50N), the desosamine moiety of both YC-17 and narbomycin was bound in a catalytically productive "buried site." This finding suggested a two-step substrate binding mechanism, whereby desosamine is recognized in the two subsites to allow the macrolide substrate to sequentially progress toward a catalytically favorable orientation. Collectively, the binding, mutagenesis, kinetic, and x-ray structural data suggest that enhancement of the catalytic activity of PikC(D50N) is due to the facilitated relocation of substrate to the buried site, which has higher binding affinity, as opposed to dissociation in solution from the transient "surface-exposed site."


Subject(s)
Amino Sugars/chemistry , Amino Sugars/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Streptomyces/enzymology , Catalysis , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/genetics , Macrolides/chemistry , Macrolides/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Streptomyces/genetics , Substrate Specificity
19.
J Biol Chem ; 283(8): 5069-80, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18089574

ABSTRACT

CYP130 is one of the 20 Mycobacterium tuberculosis cytochrome P450 enzymes, only two of which, CYP51 and CYP121, have so far been studied as individually expressed proteins. Here we characterize a third heterologously expressed M. tuberculosis cytochrome P450, CYP130, by UV-visible spectroscopy, isothermal titration calorimetry, and x-ray crystallography, including determination of the crystal structures of ligand-free and econazole-bound CYP130 at a resolution of 1.46 and 3.0A(,) respectively. Ligand-free CYP130 crystallizes in an "open" conformation as a monomer, whereas the econazole-bound form crystallizes in a "closed" conformation as a dimer. Conformational changes enabling the "open-closed" transition involve repositioning of the BC-loop and the F and G helices that envelop the inhibitor in the binding site and reshape the protein surface. Crystal structure analysis shows that the portion of the BC-loop relocates as much as 18A between the open and closed conformations. Binding of econazole to CYP130 involves a conformational change and is mediated by both a set of hydrophobic interactions with amino acid residues in the active site and coordination of the heme iron. CYP130 also binds miconazole with virtually the same binding affinity as econazole and clotrimazole and ketoconazole with somewhat lower affinities, which makes it a plausible target for this class of therapeutic drugs. Overall, binding of the azole inhibitors is a sequential two-step, entropy-driven endothermic process. Binding of econazole and clotrimazole exhibits positive cooperativity that may reflect a propensity of CYP130 to associate into a dimeric structure.


Subject(s)
Antifungal Agents/chemistry , Bacterial Proteins/chemistry , Clotrimazole/chemistry , Cytochrome P-450 Enzyme System/chemistry , Econazole/chemistry , Mycobacterium tuberculosis/enzymology , Antifungal Agents/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Clotrimazole/metabolism , Crystallography, X-Ray , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dimerization , Econazole/metabolism , Entropy , Hydrophobic and Hydrophilic Interactions , Mycobacterium tuberculosis/genetics , Protein Binding , Protein Structure, Quaternary , Spectrophotometry, Ultraviolet
20.
Antimicrob Agents Chemother ; 51(11): 3915-23, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17846131

ABSTRACT

Sterol 14alpha-demethylase (CYP51), a major checkpoint in membrane sterol biosynthesis, is a key target for fungal antibiotic therapy. We sought small organic molecules for lead candidate CYP51 inhibitors. The changes in CYP51 spectral properties following ligand binding make CYP51 a convenient target for high-throughput screening technologies. These changes are characteristic of either substrate binding (type I) or inhibitor binding (type II) in the active site. We screened a library of 20,000 organic molecules against Mycobacterium tuberculosis CYP51 (CYP51(Mt)), examined the top type I and type II binding hits for their inhibitory effects on M. tuberculosis in broth culture, and analyzed them spectrally for their ability to discriminate between CYP51(Mt) and two reference M. tuberculosis CYP proteins, CYP130 and CYP125. We determined the binding mode for one of the top type II hits, alpha-ethyl-N-4-pyridinyl-benzeneacetamide (EPBA), by solving the X-ray structure of the CYP51(Mt)-EPBA complex to a resolution of 1.53 A. EPBA binds coordinately to the heme iron in the CYP51(Mt) active site through a lone pair of nitrogen electrons and also through hydrogen bonds with residues H259 and Y76, which are invariable in the CYP51 family, and hydrophobic interactions in a phylum- and/or substrate-specific cavity of CYP51. We also identified a second compound with structural and binding properties similar to those of EPBA, 2-(benzo[d]-2,1,3-thiadiazole-4-sulfonyl)-2-amino-2-phenyl-N-(pyridinyl-4)-acetamide (BSPPA). The congruence between the geometries of EPBA and BSPPA and the CYP51 binding site singles out EPBA and BSPPA as lead candidate CYP51 inhibitors with optimization potential for efficient discrimination between host and pathogen enzymes.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzeneacetamides/chemistry , Benzeneacetamides/pharmacology , Binding Sites/genetics , Crystallography, X-Ray/methods , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Imidazoles/pharmacology , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/genetics , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...