Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters










Publication year range
1.
Drug Metab Rev ; 51(2): 162-177, 2019 05.
Article in English | MEDLINE | ID: mdl-31203694

ABSTRACT

The oxidation of carbon-carbon triple bonds by cytochrome P450 produces ketene metabolites that are hydrolyzed to acetic acid derivatives or are trapped by nucleophiles. In the special case of 17α-ethynyl sterols, D-ring expansion and de-ethynylation have been observed as competing pathways. The oxidation of acetylenic groups is also associated with mechanism-based inactivation of cytochrome P450 enzymes. One mechanism for this inactivation is reaction of the ketene metabolite with cytochrome P450 residues essential for substrate binding or catalysis. However, in the case of monosubstituted acetylenes, inactivation can also occur by addition of the oxidized acetylenic function to a nitrogen of the heme prosthetic group. This addition reaction is not mediated by the ketene metabolite, but rather occurs during oxygen transfer to the triple bond. In some instances, a detectable intermediate is formed that is most consistent with a ketocarbene-iron heme complex. This complex can progress to the N-alkylated heme or revert back to the unmodified enzyme. The ketocarbene complex may intervene in the formation of all the N-alkyl heme adducts, but is normally too unstable to be detected.


Subject(s)
Alkynes/metabolism , Cytochrome P-450 Enzyme System/metabolism , Animals , Enzyme Activation , Humans , Oxidation-Reduction
2.
Drug Metab Dispos ; 46(8): 1091-1095, 2018 08.
Article in English | MEDLINE | ID: mdl-29777023

ABSTRACT

Ethionamide (ETH) plays a central role in the treatment of tuberculosis in patients resistant to the first-line drugs. The ETH, thioamide, and thiourea class of antituberculosis agents are prodrugs that are oxidatively converted to their active S-oxides by the mycobacterial flavin-dependent monooxygenase (EtaA) of Mycobacterium tuberculosis, thus initiating the chain of reactions that result in inhibition of mycolic acid biosynthesis and cell lysis. As part of a search for new lead candidates, we report here that several xanthates are oxidized by purified EtaA to S-oxide metabolites (perxanthates), which are implicated in the antimycobacterial activity of these compounds. This process, which is analogous to that responsible for activation of ETH, is also catalyzed by human flavoprotein monooxygenase 3. EtaA was not inhibited in a time-dependent manner during the reaction. Xanthates with longer alkyl chains were oxidized more efficiently. EtaA oxidized octyl-xanthate (Km = 5 µM; Vmax = 1.023 nmolP/min; kcat = 5.2 molP/min/molE) more efficiently than ETH (194 µM; 1.46 nmolP/min; 7.73 nmolP/min/molE, respectively). Furthermore, the in vitro antimycobacterial activity of four xanthates against M. tuberculosis H37Hv was higher (minimum inhibitory concentration of around 1 µM) than that of ETH (12 µM).


Subject(s)
Anti-Bacterial Agents/metabolism , Antitubercular Agents/metabolism , Ethionamide/metabolism , Flavoproteins/metabolism , Mixed Function Oxygenases/metabolism , Anti-Bacterial Agents/pharmacology , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Ethionamide/pharmacology , Humans , Microbial Sensitivity Tests/methods , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction/drug effects , Oxygenases/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology
3.
J Inorg Biochem ; 180: 235-245, 2018 03.
Article in English | MEDLINE | ID: mdl-29352597

ABSTRACT

The Mycobacterium tuberculosis genome encodes twenty cytochrome P450 enzymes, most or all of which appear to have specific physiological functions rather than being devoted to the removal of xenobiotics. However, in many cases their specific functions remain obscure. Considerable spectroscopic, biophysical, crystallographic, and catalytic information is available on nine of these cytochrome P450 enzymes, although gaps exist in our knowledge of even these enzymes. The available evidence indicates that at least three of the better-characterized enzymes are promising targets for antituberculosis drug discovery. This review summarizes the information on the nine relatively well-characterized cytochrome P450 enzymes, with a particular emphasis on CYP121, CYP125, and CYP142 from Mycobacterium tuberculosis and Mycobacterium smegmatis.


Subject(s)
Antitubercular Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/drug effects , Mycobacterium tuberculosis/drug effects , Azoles/pharmacology , Catalysis , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Drug Discovery , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Protein Conformation
5.
Arch Biochem Biophys ; 612: 1-8, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27729224

ABSTRACT

DosS is a sensor in Mycobacterium tuberculosis that differentially responds to O2, NO, and CO, as well as to changes in the redox state of the prosthetic heme iron atom. The ferrous protein and its Fe(II)NO and Fe(II)CO complexes undergo autophosphorylation and subsequently transfer the phosphate group to DosR, a nuclear factor, to activate it. In contrast, autophosphorylation is negligible with the ferric protein and the Fe(II)O2 complex. To clarify the basis for this differential response to gases, we have determined the crystal structures of the NO and COcomplexes of the DosS GAF-A domain, which contains the heme to which the gases bind. Comparison of these crystal structures with those reported for the phosphorylation-inactive ferric GAF-A domain suggest that the GAF-A domain is in a dynamic equilibrium between active and inactive states, and that the position of Glu87 in the heme cavity, which depends on the which gas is bound, acts as a modulator of the equilibrium, and therefore of catalytic activity.


Subject(s)
Bacterial Proteins/chemistry , Carbon Monoxide/chemistry , Iron/chemistry , Mycobacterium tuberculosis/chemistry , Nitric Oxide/chemistry , Oxygen/chemistry , Protamine Kinase/chemistry , Amino Acids/chemistry , Catalysis , Crystallography, X-Ray , Heme/chemistry , Hydrogen Bonding , Oxidation-Reduction , Phosphorylation , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Signal Transduction
6.
J Biol Chem ; 291(31): 16100-11, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27235395

ABSTRACT

Mycobacterium tuberculosis DosS is critical for the induction of M. tuberculosis dormancy genes in response to nitric oxide (NO), carbon monoxide (CO), or hypoxia. These environmental stimuli, which are sensed by the DosS heme group, result in autophosphorylation of a DosS His residue, followed by phosphotransfer to an Asp residue of the response regulator DosR. To clarify the mechanism of gaseous ligand recognition and signaling, we investigated the hydrogen-bonding interactions of the iron-bound CO and NO ligands by site-directed mutagenesis of Glu-87 and His-89. Autophosphorylation assays and molecular dynamics simulations suggest that Glu-87 has an important role in ligand recognition, whereas His-89 is essential for signal transduction to the kinase domain, a process for which Arg-204 is important. Mutation of Glu-87 to Ala or Gly rendered the protein constitutively active as a kinase, but with lower autophosphorylation activity than the wild-type in the Fe(II) and the Fe(II)-CO states, whereas the E87D mutant had little kinase activity except for the Fe(II)-NO complex. The H89R mutant exhibited attenuated autophosphorylation activity, although the H89A and R204A mutants were inactive as kinases, emphasizing the importance of these residues in communication to the kinase core. Resonance Raman spectroscopy of the wild-type and H89A mutant indicates the mutation does not alter the heme coordination number, spin state, or porphyrin deformation state, but it suggests that interdomain interactions are disrupted by the mutation. Overall, these results confirm the importance of the distal hydrogen-bonding network in ligand recognition and communication to the kinase domain and reveal the sensitivity of the system to subtle differences in the binding of gaseous ligands.


Subject(s)
Bacterial Proteins , Carbon Monoxide , Mycobacterium tuberculosis , Nitric Oxide , Protamine Kinase , Signal Transduction/physiology , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon Monoxide/chemistry , Carbon Monoxide/metabolism , Hydrogen Bonding , Mutation, Missense , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Phosphorylation , Protamine Kinase/chemistry , Protamine Kinase/genetics , Protamine Kinase/metabolism
7.
Drug Metab Dispos ; 44(5): 771-80, 2016 May.
Article in English | MEDLINE | ID: mdl-26936974

ABSTRACT

CYP2W1 is a recently discovered human cytochrome P450 enzyme with a distinctive tumor-specific expression pattern. We show here that CYP2W1 exhibits tight binding affinities for retinoids, which have low nanomolar binding constants, and much poorer binding constants in the micromolar range for four other ligands. CYP2W1 converts all-transretinoic acid (atRA) to 4-hydroxyatRA and all-transretinol to 4-OH all-transretinol, and it also oxidizes retinal. The enzyme much less efficiently oxidizes 17ß-estradiol to 2-hydroxy-(17ß)-estradiol and farnesol to a monohydroxylated product; arachidonic acid is, at best, a negligible substrate. These findings indicate that CYP2W1 probably plays an important role in localized retinoid metabolism that may be intimately linked to its involvement in tumor development.


Subject(s)
Cytochrome P450 Family 2/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Arachidonic Acid/metabolism , Catalysis , Estradiol/metabolism , Humans , Ligands , Oxidation-Reduction , Protein Binding , Retinoids/metabolism , Tretinoin/metabolism
8.
J Biol Chem ; 291(14): 7325-33, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26833565

ABSTRACT

Cholest-4-en-3-one, whether added exogenously or generated intracellularly from cholesterol, inhibits the growth ofMycobacterium tuberculosiswhen CYP125A1 and CYP142A1, the cytochrome P450 enzymes that initiate degradation of the sterol side chain, are disabled. Here we demonstrate that a 16-hydroxy derivative of cholesterol, which was previously reported to inhibit growth ofM. tuberculosis, acts by preventing the oxidation of the sterol side chain even in the presence of the relevant cytochrome P450 enzymes. The finding that (25R)-cholest-5-en-3ß,16ß,26-triol (1) (and its 3-keto metabolite) inhibit growth suggests that cholesterol analogs with non-degradable side chains represent a novel class of anti-mycobacterial agents. In accord with this, two cholesterol analogs with truncated, fluorinated side chains have been synthesized and shown to similarly block the growth in culture ofM. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Cholestenones/pharmacology , Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Mycobacterium tuberculosis/growth & development , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Cholestenones/chemistry , Cholesterol/chemistry , Cytochrome P-450 Enzyme System/metabolism
9.
Biochemistry ; 54(46): 6909-16, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26522442

ABSTRACT

Mycobacterium tuberculosis (Mtb) and Mycobacterium smegmatis (Msmeg) can grow on cholesterol as the sole carbon source. In Mtb the utilization of cholesterol can be initiated by CYP125A1 or CYP142A1 and in Msmeg by the orthologous CYP125A3 and CYP142A2. Double knockout of the two enzymes in Mtb prevents its growth on cholesterol, but the double knockout of Msmeg is still able to grow, albeit at a slower rate. We report here that Msmeg has a third enzyme, CYP125A4, that also oxidizes cholesterol, although it has a much higher activity for the oxidation of 7α-hydroxycholesterol. The ability of Msmeg CYP125A4 (and Mtb CYP125A1) to oxidize 7α-hydroxycholesterol is due, at least in part, to the presence of a smaller amino acid side chain facing C-7 of the sterol substrate than in CYP125A3. The ability to oxidize 7-substituted steroids broadens the range of sterol carbon sources for growth, but even more importantly in Mtb, additional biological effects are possible due to the potent immunomodulatory activity of 7α,26-dihydroxycholesterol.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cholesterol 7-alpha-Hydroxylase/chemistry , Cholesterol 7-alpha-Hydroxylase/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium smegmatis/enzymology , Amino Acid Substitution , Bacterial Proteins/genetics , Catalytic Domain , Cholesterol 7-alpha-Hydroxylase/genetics , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/genetics , Gene Knockout Techniques , Genes, Bacterial , Hydroxycholesterols/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/growth & development , Spectrophotometry , Structural Homology, Protein , Substrate Specificity
11.
J Biol Chem ; 289(44): 30417-30425, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25210044

ABSTRACT

Mycobacteria share a common cholesterol degradation pathway initiated by oxidation of the alkyl side chain by enzymes of cytochrome P450 (CYP) families 125 and 142. Structural and sequence comparisons of the two enzyme families revealed two insertions into the N-terminal region of the CYP125 family (residues 58-67 and 100-109 in the CYP125A1 sequence) that could potentially sterically block the oxidation of the longer cholesterol ester molecules. Catalytic assays revealed that only CYP142 enzymes are able to oxidize cholesteryl propionate, and although CYP125 enzymes could oxidize cholesteryl sulfate, they were much less efficient at doing so than the CYP142 enzymes. The crystal structure of CYP142A2 in complex with cholesteryl sulfate revealed a substrate tightly fit into a smaller active site than was previously observed for the complex of CYP125A1 with 4-cholesten-3-one. We propose that the larger CYP125 active site allows for multiple binding modes of cholesteryl sulfate, the majority of which trigger the P450 catalytic cycle, but in an uncoupled mode rather than one that oxidizes the sterol. In contrast, the more unhindered and compact CYP142 structure enables enzymes of this family to readily oxidize cholesteryl esters, thus providing an additional source of carbon for mycobacterial growth.


Subject(s)
Cholesterol Esters/chemistry , Cytochrome P-450 Enzyme System/chemistry , Mycobacterium smegmatis/enzymology , Catalytic Domain , Crystallography, X-Ray , Kinetics , Models, Molecular , Mycobacterium tuberculosis/enzymology , NADP/chemistry , Oxidation-Reduction , Protein Binding , Protein Structure, Secondary
12.
Drug Metab Dispos ; 42(5): 828-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24584631

ABSTRACT

Carbon-carbon bond cleavage reactions are catalyzed by, among others, lanosterol 14-demethylase (CYP51), cholesterol side-chain cleavage enzyme (CYP11), sterol 17ß-lyase (CYP17), and aromatase (CYP19). Because of the high substrate specificities of these enzymes and the complex nature of their substrates, these reactions have been difficult to characterize. A CYP1A2-catalyzed carbon-carbon bond cleavage reaction is required for conversion of the prodrug nabumetone to its active form, 6-methoxy-2-naphthylacetic acid (6-MNA). Despite worldwide use of nabumetone as an anti-inflammatory agent, the mechanism of its carbon-carbon bond cleavage reaction remains obscure. With the help of authentic synthetic standards, we report here that the reaction involves 3-hydroxylation, carbon-carbon cleavage to the aldehyde, and oxidation of the aldehyde to the acid, all catalyzed by CYP1A2 or, less effectively, by other P450 enzymes. The data indicate that the carbon-carbon bond cleavage is mediated by the ferric peroxo anion rather than the ferryl species in the P450 catalytic cycle. CYP1A2 also catalyzes O-demethylation and alcohol to ketone transformations of nabumetone and its analogs.


Subject(s)
Butanones/metabolism , Cyclooxygenase 2 Inhibitors/metabolism , Cytochrome P-450 CYP1A2/metabolism , Naphthaleneacetic Acids/metabolism , Prodrugs/metabolism , Biocatalysis , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/genetics , Escherichia coli/genetics , Humans , In Vitro Techniques , Microsomes/enzymology , Microsomes/metabolism , Nabumetone , Oxidation-Reduction , Substrate Specificity , Transfection
13.
Mol Pharmacol ; 84(3): 451-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23821647

ABSTRACT

Cytochrome P450 (P450)-catalyzed oxidation of the aromatic ring of estradiol can result in 2- or 4-hydroxylation. Which of these products is formed is biologically important, as the 4-hydroxylated metabolite is carcinogenic, whereas the 2-hydroxylated metabolite is not. Most human P450 enzymes, including CYP1A1 and CYP1A2, exhibit a high preference for estradiol 2-hydroxylation, but human CYP1B1 greatly favors 4-hydroxylation. Here we show that heterologous expression of the human, monkey, dog, rat, and mouse CYP1B1 enzymes yields active proteins that differ in their estradiol hydroxylation specificity. The monkey and dog orthologs, like the human enzyme, preferentially catalyze 4-hydroxylation, but the rat and mouse enzymes favor 2-hydroxylation. Analysis of the CYP1B1 sequences in light of these findings suggested that one residue, Val395 in human CYP1B1, could account for the differential hydroxylation specificities. In fact, mutation of this valine in human CYP1B1 to the leucine present in the rat enzyme produces a human enzyme that has the 2-hydroxylation specificity of the rat enzyme. The converse is true when the leucine in the rat enzyme is mutated to the human valine. The role of CYP1B1 in estradiol carcinogenicity thus depends on the identity of this single amino acid residue.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Estradiol/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP1B1 , Dogs , Humans , Hydroxylation , Macaca mulatta , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , NADPH-Ferrihemoprotein Reductase/metabolism , Rats , Species Specificity
14.
Environ Microbiol ; 15(8): 2342-59, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23489718

ABSTRACT

Degradation of the cholesterol side-chain in Mycobacterium tuberculosis is initiated by two cytochromes P450, CYP125A1 and CYP142A1, that sequentially oxidize C26 to the alcohol, aldehyde and acid metabolites. Here we report characterization of the homologous enzymes CYP125A3 and CYP142A2 from Mycobacterium smegmatis mc(2) 155. Heterologously expressed, purified CYP125A3 and CYP142A2 bound cholesterol, 4-cholesten-3-one, and antifungal azole drugs. CYP125A3 or CYP142A2 reconstituted with spinach ferredoxin and ferredoxin reductase efficiently hydroxylated 4-cholesten-3-one to the C-26 alcohol and subsequently to the acid. The X-ray structures of both substrate-free CYP125A3 and CYP142A2 and of cholest-4-en-3-one-bound CYP142A2 reveal significant differences in the substrate binding sites compared with the homologous M. tuberculosis proteins. Deletion only of cyp125A3 causes a reduction of both the alcohol and acid metabolites and a strong induction of cyp142 at the mRNA and protein levels, indicating that CYP142A2 serves as a functionally redundant back up enzyme for CYP125A3. In contrast to M. tuberculosis, the M. smegmatis Δcyp125Δcyp142 double mutant retains its ability to grow on cholesterol albeit with a diminished capacity, indicating an additional level of redundancy within its genome.


Subject(s)
Cholesterol/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/genetics , Antifungal Agents/metabolism , Azoles/metabolism , Binding Sites , Cholestenones/metabolism , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/chemistry , Gene Deletion , Gene Expression Regulation, Bacterial , Hydroxylation , Models, Molecular , Mycobacterium smegmatis/growth & development , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Oxidation-Reduction , Protein Structure, Tertiary
15.
Future Med Chem ; 5(2): 213-28, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23360144

ABSTRACT

A prodrug is a compound that has negligible, or lower, activity against a specified pharmacological target than one of its major metabolites. Prodrugs can be used to improve drug delivery or pharmacokinetics, to decrease toxicity, or to target the drug to specific cells or tissues. Ester and phosphate hydrolysis are widely used in prodrug design because of their simplicity, but such approaches are relatively ineffective for targeting drugs to specific sites. The activation of prodrugs by the cytochrome P450 system provides a highly versatile approach to prodrug design that is particularly adaptable for targeting drug activation to the liver, to tumors or to hypoxic tissues.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Delivery Systems , Prodrugs/chemistry , Prodrugs/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Drug Delivery Systems/methods , Humans , Prodrugs/administration & dosage
16.
J Biol Chem ; 288(10): 7147-57, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23362256

ABSTRACT

Intramolecular disulfide bond formation is promoted in oxidizing extracellular and endoplasmic reticulum compartments and often contributes to protein stability and function. DUOX1 and DUOX2 are distinguished from other members of the NOX protein family by the presence of a unique extracellular N-terminal region. These peroxidase-like domains lack the conserved cysteines that confer structural stability to mammalian peroxidases. Sequence-based structure predictions suggest that the thiol groups present are solvent-exposed on a single protein surface and are too distant to support intramolecular disulfide bond formation. To investigate the role of these thiol residues, we introduced four individual cysteine to glycine mutations in the peroxidase-like domains of both human DUOXs and purified the recombinant proteins. The mutations caused little change in the stabilities of the monomeric proteins, supporting the hypothesis that the thiol residues are solvent-exposed and not involved in disulfide bonds that are critical for structural integrity. However, the ability of the isolated hDUOX1 peroxidase-like domain to dimerize was altered, suggesting a role for these cysteines in protein-protein interactions that could facilitate homodimerization of the peroxidase-like domain or, in the full-length protein, heterodimeric interactions with a maturation protein. When full-length hDUOX1 was expressed in HEK293 cells, the mutations resulted in decreased H2O2 production that correlated with a decreased amount of the enzyme localized to the membrane surface rather than with a loss of activity or with a failure to synthesize the mutant proteins. These results support a role for the cysteine residues in intermolecular disulfide bond formation with the DUOX maturation factor DUOXA1.


Subject(s)
Cysteine/metabolism , Membrane Proteins/metabolism , NADPH Oxidases/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , Cysteine/chemistry , Cysteine/genetics , Dual Oxidases , Electrophoresis, Polyacrylamide Gel , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutation , NADPH Oxidases/chemistry , NADPH Oxidases/genetics , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Sf9 Cells , Spectrometry, Fluorescence , Surface Properties
17.
Biochemistry ; 51(37): 7403-16, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22946928

ABSTRACT

We have recombinantly expressed and purified the ligand binding domains (LBDs) of four insect nuclear receptors of the E75 family. The Drosophila melanogaster and Bombyx mori nuclear receptors were purified as ferric hemoproteins with Soret maxima at 424 nm, whereas their ferrous forms had a Soret maximum at 425 nm that responds to (•)NO and CO binding. In contrast, the purified LBD of Oncopeltus fasciatus displayed a Soret maximum at 415 nm for the ferric protein that shifted to 425 nm in its ferrous state. Binding of (•)NO to the heme moiety of the D. melanogaster and B. mori E75 LBD resulted in the appearance of a peak at 385 nm, whereas this peak appeared at 416 nm in the case of the O. fasciatus hemoprotein, resembling the behavior displayed by its human homologue, Rev-erbß. High-performance liquid chromatography analysis revealed that, unlike the D. melanogaster and B. mori counterparts, the heme group of O. fasciatus is covalently attached to the protein through the side chains of two amino acids. The high degree of sequence homology with O. fasciatus E75 led us to clone and express the LBD of Blattella germanica, which established that its spectral properties closely resemble those of O. fasciatus and that it also has the heme group covalently bound to the protein. Hence, (•)NO/CO regulation of the transcriptional activity of these nuclear receptors might be differently controlled among various insect species. In addition, covalent heme binding provides strong evidence that at least some of these nuclear receptors function as diatomic gas sensors rather than heme sensors. Finally, our findings expand the classes of hemoproteins in which the heme group is normally covalently attached to the polypeptide chain.


Subject(s)
DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Heme/chemistry , Insect Proteins/chemistry , Nitric Oxide/chemistry , Receptors, Steroid/chemistry , Animals , Bombyx , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Heme/genetics , Heme/metabolism , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Nitric Oxide/genetics , Nitric Oxide/metabolism , Protein Structure, Tertiary , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sequence Homology, Amino Acid , Species Specificity
18.
J Biol Chem ; 287(42): 35181-35191, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22923613

ABSTRACT

Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. HO-1 is an inducible enzyme, but HO-2 is constitutively expressed in selected tissues and is involved in signaling and regulatory processes. HO-2 has three cysteine residues that have been proposed to modulate the affinity for heme, whereas HO-1 has none. Here we use site-specific mutagenesis and two-dimensional NMR of l-[3-(13)C]cysteine-labeled proteins to determine the redox state of the individual cysteines in HO-2 and assess their roles in binding of heme. The results indicate that in the apoprotein, Cys(282) and Cys(265) are in the oxidized state, probably in an intramolecular disulfide bond. The addition of a reducing agent converts them to the reduced, free thiol state. Two-dimensional NMR of site-specific mutants reveals that the redox state of Cys(265) and Cys(282) varies with the presence or absence of other Cys residues, indicating that the microenvironments of the Cys residues are mutually interdependent. Cys(265) appears to be in a relatively hydrophilic, oxidizable environment compared with Cys(127) and Cys(282). Chemical shift data indicate that none of the cysteines stably coordinates to the heme iron atom. In the oxidized state of the apoprotein, heme is bound 2.5-fold more tightly than in the reduced state. This small difference in heme affinity between the oxidized and reduced states of the protein is much less than previously reported, suggesting that it is not a significant factor in the physiological regulation of cellular heme levels.


Subject(s)
Cysteine/chemistry , Heme Oxygenase (Decyclizing)/chemistry , Heme/chemistry , Protein Folding , Carbon Isotopes/chemistry , Cysteine/genetics , Cysteine/metabolism , Heme/genetics , Heme/metabolism , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Humans , Isotope Labeling , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Protein Binding , Protein Structure, Tertiary
19.
Bioorg Med Chem ; 20(13): 4064-81, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22647881

ABSTRACT

We report the synthesis and evaluation of a series of cholesterol side-chain analogs as mechanistic probes of three important Mycobacterium tuberculosis cytochrome P450 enzymes that selectively oxidize the ω-position of the methyl-branched cholesterol side-chain. To probe the structural requirements for the thermodynamically disfavored ω-regiospecificity we compared the binding of these substrate analogs to each P450, determined the turnover rates, and characterized the enzymatic products. The results are discussed in the context of the structure-activity relationships of the enzymes and how their active sites enforce ω-oxidation.


Subject(s)
Cholesterol/metabolism , Cytochrome P-450 Enzyme System/metabolism , Mycobacterium tuberculosis/enzymology , Biocatalysis , Catalytic Domain , Cholesterol/analogs & derivatives , Cholesterol/chemical synthesis , Oxidation-Reduction , Protein Binding , Protein Isoforms/metabolism , Stereoisomerism , Substrate Specificity
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...