Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biochemistry ; 50(19): 3968-74, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21446712

ABSTRACT

Human cytochrome P450c21 (steroid 21-hydroxylase, CYP21A2) catalyzes the 21-hydroxylation of progesterone (P4) and its preferred substrate 17α-hydroxyprogestrone (17OHP4). CYP21A2 activities, which are required for cortisol and aldosterone biosynthesis, involve the formation of energetically disfavored primary carbon radicals. Therefore, we hypothesized that the binding of P4 and 17OHP4 to CYP21A2 restricts access of the reactive heme-oxygen complex to the C-21 hydrogen atoms, suppressing oxygenation at kinetically more favorable sites such as C-17 and C-16, which are both hydroxylated by cytochrome P450c17 (CYP17A1). We reasoned that expansion of the CYP21A2 substrate-binding pocket would increase substrate mobility and might yield additional hydroxylation activities. We built a computer model of CYP21A2 based principally on the crystal structure of CYP2C5, which also 21-hydroxylates P4. Molecular dynamics simulations indicate that binding of the steroid nucleus perpendicular to the plane of the CYP21A2 heme ring limits access of the heme oxygen to the C-21 hydrogen atoms. Residues L107, L109, V470, I471, and V359 were found to contribute to the CYP21A2 substate-binding pocket. Mutation of V470 and I471 to alanine or glycine preserved P4 21-hydroxylase activity, and mutations of L107 or L109 were inactive. Mutations V359A and V359G, in contrast, acquired 16α-hydroxylase activity, accounting for 40% and 90% of the P4 metabolites, respectively. We conclude that P4 binds to CYP21A2 in a fundamentally different orientation than to CYP17A1 and that expansion of the CYP21A2 substrate-binding pocket allows additional substrate trajectories and metabolic switching.


Subject(s)
Progesterone/analogs & derivatives , Steroid 21-Hydroxylase/chemistry , Amino Acid Sequence , Amino Acid Substitution/genetics , Computer Simulation , HEK293 Cells , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Progesterone/chemistry , Progesterone/genetics , Steroid 21-Hydroxylase/genetics , Substrate Specificity/genetics
2.
Arch Biochem Biophys ; 418(2): 151-60, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14522586

ABSTRACT

The 5alpha-reduction of testosterone in target tissues is a key step in androgen physiology; however, 5alpha-reduced C(19) steroids are sometimes synthesized in testis via a pathway that does not involve testosterone as an intermediate. We studied the metabolism of 5alpha-reduced C(21) steroids by human cytochrome P450c17 (hCYP17), the enzyme responsible for conversion of C(21) steroids to C(19) steroids via its 17alpha-hydroxylase and 17,20-lyase activities. hCYP17 17alpha-hydroxylates 5alpha-pregnan-3,20-dione, but little androstanedione is formed by 17,20-lyase activity. hCYP17 also 17alpha-hydroxylates 5alpha-pregnan-3alpha-ol-20-one and the 5alpha-pregnan-3alpha,17alpha-diol-20-one intermediate is rapidly converted to androsterone by 17,20-lyase activity. Furthermore, 5alpha-pregnan-3alpha,17alpha-diol-20-one is a better substrate for the 17,20-lyase reaction than the preferred substrate 17alpha-hydroxypregnenolone and cytochrome b(5) stimulates androsterone formation only 3-fold. Both 5alpha-pregnan-3alpha-ol-20-one and 5alpha-pregnan-3alpha,17alpha-diol-20-one bind to hCYP17 with higher affinity than does progesterone. We conclude that 5alpha-reduced, 3alpha-hydroxy-C(21) steroids are excellent, high-affinity substrates for hCYP17. The brisk metabolism of 5alpha-pregnan-3alpha,17alpha-diol-20-one to androsterone by CYP17 explains how, when 5alpha-reductases are present, the testis can produce C(19) steroids androsterone and androstanediol from 17alpha-hydroxyprogesterone without the intermediacy of androstenedione and testosterone.


Subject(s)
5-alpha-Dihydroprogesterone/chemistry , 5-alpha-Dihydroprogesterone/metabolism , Androgens/biosynthesis , Pregnanolone/chemistry , Pregnanolone/metabolism , Steroid 17-alpha-Hydroxylase/chemistry , Steroid 17-alpha-Hydroxylase/metabolism , Gonads/metabolism , Humans , Hydroxylation , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Steroids/chemistry , Steroids/metabolism , Substrate Specificity
3.
Arch Biochem Biophys ; 409(1): 134-44, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12464252

ABSTRACT

Human cytochrome P450c17 (17alpha-hydroxylase, 17,20-lyase) (CYP17) and cytochrome P450c21 (21-hydroxylase) (CYP21) differ by only 14 amino acids in length and share 29% amino acid identity. Both enzymes hydroxylate progesterone at carbon atoms that lie only 2.6A apart, but CYP17 also metabolizes other steroids and demonstrates additional catalytic activities. To probe the active site topologies of these related enzymes, we synthesized the enantiomer of progesterone and determined if ent-progesterone is a substrate or inhibitor of CYP17 and CYP21. Neither enzyme metabolizes ent-progesterone; however, ent-progesterone is a potent competitive inhibitor of CYP17 (K(I)=0.2 microM). The ent-progesterone forms a type I difference spectrum with CYP17, but molecular dynamics simulations suggest different binding orientations for progesterone and its enantiomer. The ent-progesterone also inhibits CYP21, with weaker affinity than for CYP17. We conclude that CYP17 accommodates the stereochemically unnatural ent-progesterone better than CYP21. Enantiomeric steroids can be used to probe steroid binding sites, and these compounds may be effective inhibitors of steroid biosynthesis.


Subject(s)
Progesterone/chemistry , Steroid 17-alpha-Hydroxylase/chemistry , Steroid 21-Hydroxylase/chemistry , Binding Sites , Binding, Competitive , Humans , Kinetics , Microsomes/metabolism , Models, Chemical , Models, Molecular , Plasmids/metabolism , Progesterone/pharmacology , Protein Binding , Saccharomyces cerevisiae/metabolism , Spectrophotometry , Stereoisomerism , Substrate Specificity , Temperature , Time Factors
4.
Endocr Res ; 28(4): 443-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12530647

ABSTRACT

Cytochrome P450c17 (CYP17) is a single hemoprotein that catalyzes both the 17alpha-hydroxylase and 17,20-lyase reactions in all species thus far examined. Severe defects in CYP17 cause classical 17-hydroxylase deficiency, but other defects result in partial or selective deficiency states. One such variant is the syndrome of isolated 17,20-lyase deficiency. Recent detailed studies of the biochemical properties of the mutant CYP17 enzymes from patients with isolated 17,20-lyase deficiency demonstrate that alterations in the interaction of CYP17 with its redox partner proteins P450-oxidoreductase and cytochrome b5 form the biochemical basis for these selective enzyme defects. Site-directed mutagenesis studies have confirmed that neutralization of any of several positive charges on the redox partner binding surface results in selective disruption of 17,20-lyase activity. In one case diagnosed as isolated 17,20-lyase deficiency, the identified mutation did not map to the redox partner binding surface; however, we have shown that this mutation cannot be the cause of isolated 17,20-lyase deficiency in this patient. These consistent results have prompted us to propose a paradigm in which neutralization of positive charges in the redox partner binding surface of CYP17 may be the predominant if not sole mechanism leading to isolated 17,20-lyase deficiency.


Subject(s)
Adrenal Hyperplasia, Congenital/enzymology , Adrenal Hyperplasia, Congenital/genetics , Mutation/physiology , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Animals , Binding Sites/physiology , COS Cells , Electrochemistry , Humans , Mutagenesis, Site-Directed , Oxidation-Reduction , Steroid 17-alpha-Hydroxylase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...