Conversion of Human Steroid 5β-Reductase (AKR1D1) into 3β-Hydroxysteroid Dehydrogenase by Single Point Mutation E120H

Background: The aldo-keto reductase (AKR) subfamily AKR1D comprises steroid 5β-reductases, whereas the AKR1C subfamily comprises hydroxysteroid dehydrogenases. Results: A single E120H mutation in AKR1D1 converts human steroid 5β-reductase into a 3β-hydroxysteroid dehydrogenase. Conclusion: Glu120 controls the positional specificity of hydride transfer and facilitates double bond reduction. Significance: This is an example of a perfect change-of-function that can be achieved by a single point mutation. Human aldo-keto reductase 1D1 (AKR1D1) and AKR1C enzymes are essential for bile acid biosynthesis and steroid hormone metabolism. AKR1D1 catalyzes the 5β-reduction of Δ4-3-ketosteroids, whereas AKR1C enzymes are hydroxysteroid dehydrogenases (HSDs). These enzymes share high sequence identity and catalyze 4-pro-(R)-hydride transfer from NADPH to an electrophilic carbon but differ in that one residue in the conserved AKR catalytic tetrad, His120 (AKR1D1 numbering), is substituted by a glutamate in AKR1D1. We find that the AKR1D1 E120H mutant abolishes 5β-reductase activity and introduces HSD activity. However, the E120H mutant unexpectedly favors dihydrosteroids with the 5α-configuration and, unlike most of the AKR1C enzymes, shows a dominant stereochemical preference to act as a 3β-HSD as opposed to a 3α-HSD. The catalytic efficiency achieved for 3β-HSD activity is higher than that observed for any AKR to date. High resolution crystal structures of the E120H mutant in complex with epiandrosterone, 5β-dihydrotestosterone, and Δ4-androstene-3,17-dione elucidated the structural basis for this functional change. The glutamate-histidine substitution prevents a 3-ketosteroid from penetrating the active site so that hydride transfer is directed toward the C3 carbonyl group rather than the Δ4-double bond and confers 3β-HSD activity on the 5β-reductase. Structures indicate that stereospecificity of HSD activity is achieved because the steroid flips over to present its α-face to the A-face of NADPH. This is in contrast to the AKR1C enzymes, which can invert stereochemistry when the steroid swings across the binding pocket. These studies show how a single point mutation in AKR1D1 can introduce HSD activity with unexpected configurational and stereochemical preference.

[1]  T. Penning,et al.  Substrate specificity and inhibitor analyses of human steroid 5β-reductase (AKR1D1) , 2011, Steroids.

[2]  Randy J. Read,et al.  Acta Crystallographica Section D Biological , 2003 .

[3]  A. Mildvan,et al.  Hydrogen Bonding at the Active Site of Δ5-3-Ketosteroid Isomerase† , 1997 .

[4]  T. Penning,et al.  Molecular docking simulations of steroid substrates into human cytosolic hydroxysteroid dehydrogenases (AKR1C1 and AKR1C2): Insights into positional and stereochemical preferences , 2006, Steroids.

[5]  Trevor M Penning,et al.  Human aldo-keto reductases: Function, gene regulation, and single nucleotide polymorphisms. , 2007, Archives of biochemistry and biophysics.

[6]  S. Granick,et al.  Stimulation of hemoglobin synthesis in chick blastoderms by certain 5beta androstane and 5beta pregnane steroids. , 1967, Proceedings of the National Academy of Sciences of the United States of America.

[7]  D. Christianson,et al.  Crystal Structure of Human Liver Δ4-3-Ketosteroid 5β-Reductase (AKR1D1) and Implications for Substrate Binding and Catalysis*♦ , 2008, Journal of Biological Chemistry.

[8]  G. Salen,et al.  Bile acid biosynthesis , 1974, The American Journal of Digestive Diseases.

[9]  L. Moore,et al.  Orphan Nuclear Receptors Constitutive Androstane Receptor and Pregnane X Receptor Share Xenobiotic and Steroid Ligands* , 2000, The Journal of Biological Chemistry.

[10]  N. Palackal,et al.  Crystal structure of human type III 3alpha-hydroxysteroid dehydrogenase/bile acid binding protein complexed with NADP(+) and ursodeoxycholate. , 2001, Biochemistry.

[11]  Sheng-Xiang Lin,et al.  Crystal structures of the multispecific 17beta-hydroxysteroid dehydrogenase type 5: critical androgen regulation in human peripheral tissues. , 2004, Molecular endocrinology.

[12]  I. Blair,et al.  Stereospecific reduction of 5β-reduced steroids by human ketosteroid reductases of the AKR (aldo-keto reductase) superfamily: role of AKR1C1-AKR1C4 in the metabolism of testosterone and progesterone via the 5β-reductase pathway. , 2011, The Biochemical journal.

[13]  J. Thornton,et al.  PROCHECK: a program to check the stereochemical quality of protein structures , 1993 .

[14]  Michael E. Burczynski,et al.  Human 3α-hydroxysteroid dehydrogenase isoforms (AKR1C1–AKR1C4) of the aldo-keto reductase superfamily: functional plasticity and tissue distribution reveals roles in the inactivation and formation of male and female sex hormones , 2000 .

[15]  J. Jez,et al.  Engineering Steroid 5β-Reductase Activity into Rat Liver 3α-Hydroxysteroid Dehydrogenase† , 1998 .

[16]  B. Oh,et al.  Crystal Structure of Δ5-3-Ketosteroid Isomerase from Pseudomonas testosteroni in Complex with Equilenin Settles the Correct Hydrogen Bonding Scheme for Transition State Stabilization* , 1999, The Journal of Biological Chemistry.

[17]  M. Lewis,et al.  Comparative anatomy of the aldo-keto reductase superfamily. , 1997, The Biochemical journal.

[18]  Seon-Hwa Lee,et al.  Human Cytosolic Hydroxysteroid Dehydrogenases of the Aldo-ketoreductase Superfamily Catalyze Reduction of Conjugated Steroids , 2009, Journal of Biological Chemistry.

[19]  B. Oh,et al.  High-resolution crystal structures of delta5-3-ketosteroid isomerase with and without a reaction intermediate analogue. , 1997, Biochemistry.

[20]  Jacques Simard,et al.  Molecular Biology of the 3-Hydroxysteroid Dehydrogenase/5-4 Isomerase Gene Family , 2005 .

[21]  W. Kreis,et al.  The Crystal Structure of Progesterone 5β-Reductase from Digitalis lanata Defines a Novel Class of Short Chain Dehydrogenases/Reductases* , 2008, Journal of Biological Chemistry.

[22]  R J Read,et al.  Crystallography & NMR system: A new software suite for macromolecular structure determination. , 1998, Acta crystallographica. Section D, Biological crystallography.

[23]  D. Christianson,et al.  Structure and catalytic mechanism of human steroid 5β-reductase (AKR1D1) , 2009, Molecular and Cellular Endocrinology.

[24]  Z. Otwinowski,et al.  Processing of X-ray diffraction data collected in oscillation mode. , 1997, Methods in enzymology.

[25]  Kevin Cowtan,et al.  research papers Acta Crystallographica Section D Biological , 2005 .

[26]  S. Brennecke,et al.  5β-Dihydroprogesterone and steroid 5β–reductase decrease in association with human parturition at term , 2005 .

[27]  F. Faucher,et al.  The crystal structure of human Delta4-3-ketosteroid 5beta-reductase defines the functional role of the residues of the catalytic tetrad in the steroid double bond reduction mechanism. , 2008, Biochemistry.

[28]  M. Massiah,et al.  Solution structure of Delta 5-3-ketosteroid isomerase complexed with the steroid 19-nortestosterone hemisuccinate. , 1999, Biochemistry.

[29]  J. Jez,et al.  Mutagenesis of 3 alpha-hydroxysteroid dehydrogenase reveals a "push-pull" mechanism for proton transfer in aldo-keto reductases. , 1998, Biochemistry.

[30]  I. Björkhem,et al.  Cloning and expression of cDNA of human Δ4‐3‐oxosteroid 5β‐reductase and substrate specificity of the expressed enzyme , 1994 .

[31]  Pierre Legrand,et al.  Structure of the Human 3α-Hydroxysteroid Dehydrogenase Type 3 in Complex with Testosterone and NADP at 1.25-Å Resolution* , 2001, The Journal of Biological Chemistry.

[32]  G. F. Woods,et al.  Partial reduction of steroid hormones and related substances , 1955 .

[33]  K. Okuda,et al.  Purification and characterization of delta 4-3-ketosteroid 5 beta-reductase. , 1984, The Journal of biological chemistry.

[34]  Randy J. Read,et al.  Phaser crystallographic software , 2007, Journal of applied crystallography.

[35]  I. Björkhem,et al.  Enzymatic Conversion of a Δ4‐3‐Ketosteroid into a 3α‐Hydroxy‐5β‐Steroid: Mechanism and Stereochemistry of Hydrogen Transfer from NADPH , 1967 .

[36]  R Ohlsson,et al.  Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[37]  T. Penning,et al.  Human cytosolic 3alpha-hydroxysteroid dehydrogenases of the aldo-keto reductase superfamily display significant 3beta-hydroxysteroid dehydrogenase activity: implications for steroid hormone metabolism and action. , 2003, The Journal of biological chemistry.

[38]  Fernand Labrie,et al.  Human 20alpha-hydroxysteroid dehydrogenase: crystallographic and site-directed mutagenesis studies lead to the identification of an alternative binding site for C21-steroids. , 2003, Journal of molecular biology.

[39]  M. J. Bennett,et al.  Steroid recognition and regulation of hormone action: crystal structure of testosterone and NADP+ bound to 3 alpha-hydroxysteroid/dihydrodiol dehydrogenase. , 1997, Structure.

[40]  Jerry March,et al.  Advanced Organic Chemistry: Reactions, Mechanisms, and Structure , 1977 .

[41]  Michael F. Summers,et al.  Solution Structure of 3-Oxo-Δ5-Steroid Isomerase , 1997 .

[42]  S. Granick,et al.  Steroid control of porphyrin and heme biosynthesis: a new biological function of steroid hormone metabolites. , 1967, Proceedings of the National Academy of Sciences of the United States of America.