Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1844(6): 1128-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24613493

ABSTRACT

Lecithin:retinol acyltransferase (LRAT) plays a major role in the vertebrate visual cycle. Indeed, it is responsible for the esterification of all-trans retinol into all-trans retinyl esters, which can then be stored in microsomes or further metabolized to produce the chromophore of rhodopsin. In the present study, a detailed characterization of the enzymatic properties of truncated LRAT (tLRAT) has been achieved using in vitro assay conditions. A much larger tLRAT activity has been obtained compared to previous reports and to an enzyme with a similar activity. In addition, tLRAT is able to hydrolyze phospholipids bearing different chain lengths with a preference for micellar aggregated substrates. It therefore presents an interfacial activation property, which is typical of classical phospholipases. Furthermore, given that stability is a very important quality of an enzyme, the influence of different parameters on the activity and stability of tLRAT has thus been studied in detail. For example, storage buffer has a strong effect on tLRAT activity and high enzyme stability has been observed at room temperature. The thermostability of tLRAT has also been investigated using circular dichroism and infrared spectroscopy. A decrease in the activity of tLRAT was observed beyond 70°C, accompanied by a modification of its secondary structure, i.e. a decrease of its α-helical content and the appearance of unordered structures and aggregated ß-sheets. Nevertheless, residual activity could still be observed after heating tLRAT up to 100°C. The results of this study highly improved our understanding of this enzyme.


Subject(s)
Acyltransferases/chemistry , Lecithins/chemistry , Phospholipid Ethers/chemistry , Vitamin A/chemistry , Acyltransferases/genetics , Circular Dichroism , Escherichia coli/genetics , Escherichia coli/metabolism , Hot Temperature , Humans , Kinetics , Micelles , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrophotometry, Infrared , Substrate Specificity
2.
Biochemistry ; 47(51): 13537-46, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19075558

ABSTRACT

The 5beta-reductases (AKR1D1-3) are unique enzymes able to catalyze efficiently and in a stereospecific manner the 5beta-reduction of the C4-C5 double bond found in Delta4-3-ketosteroids, including steroid hormones and bile acids precursors such as 7alpha-hydroxy-4-cholesten-3-one and 7alpha,12alpha-dihydroxy-4-cholesten-3-one. In order to elucidate the binding mode and substrate specificity in detail, biochemical and structural studies on human 5beta-reductase (h5beta-red; AKR1D1) have been recently undertaken. The crystal structure of a h5beta-red binary complex provides a complete picture of the NADPH-enzyme interactions involving the flexible loop B, which contributes to the maintenance of the cofactor in its binding site by acting as a "safety belt". Structural comparison with binary complexes of AKR1C enzymes, specifically the human type 3 3alpha-hydroxysteroid dehydrogenase (AKR1C2) and the mouse 17alpha-hydroxysteroid dehydrogenase (AKR1C21), also revealed particularities in loop B positioning that make the steroid-binding cavity of h5beta-red substantially larger than those of the two other enzymes. Kinetic characterization of the purified recombinant h5beta-red has shown that this enzyme exerts a strong activity toward progesterone (Prog) and androstenedione (Delta4) but is rapidly inhibited by these substrates once their concentrations reach 2-times their K(m) value. A crystal structure of the h5beta-red in ternary complex with NADPH and Delta4 has revealed that the large steroid-binding site of this enzyme also contains a subsite in which the Delta4 molecule is found. When bound in this subsite, Delta4 completely impedes the passage of another substrate molecule toward the catalytic site. The importance of this alternative binding site for the inhibition of h5beta-red was finally proven by site-directed mutagenesis, which demonstrated that the replacement of one of the residues delineating this site (Val(309)) by a phenylalanine completely abolishes the substrate inhibition. The results of this report provide structural insights into the substrate inhibition of h5beta-red by C19- and C21-steroids.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/chemistry , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Kinetics , Molecular Conformation , Mutagenesis, Site-Directed , NADP/chemistry , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Substrate Specificity
3.
Biochemistry ; 47(32): 8261-70, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18624455

ABSTRACT

The 5beta-reductases (AKR1D1-3) are unique enzymes able to catalyze efficiently and in a stereospecific manner the 5beta-reduction of the C4-C5 double bond found into Delta4-3-ketosteroids, including steroid hormones and bile acids. Multiple-sequence alignments and mutagenic studies have already identified one of the residues presumably located at their active site, Glu 120, as the major molecular determinant for the unique activity displayed by 5beta-reductases. To define the exact role played by this glutamate in the catalytic activity of these enzymes, biochemical and structural studies on human 5beta-reductase (h5beta-red) have been undertaken. The crystal structure of h5beta-red in a ternary complex with NADP (+) and 5beta-dihydroprogesterone (5beta-DHP), the product of the 5beta-reduction of progesterone (Prog), revealed that Glu 120 does not interact directly with the other catalytic residues, as previously hypothesized, thus suggesting that this residue is not directly involved in catalysis but could instead be important for the proper positioning of the steroid substrate in the catalytic site. On the basis of our structural results, we thus propose a realistic scheme for the catalytic mechanism of the C4-C5 double bond reduction. We also propose that bile acid precursors such as 7alpha-hydroxy-4-cholesten-3-one and 7alpha,12alpha-dihydroxy-4-cholesten-3-one, when bound to the active site of h5beta-red, can establish supplementary contacts with Tyr 26 and Tyr 132, two residues delineating the steroid-binding cavity. These additional contacts very likely account for the higher activity of h5beta-red toward the bile acid intermediates versus steroid hormones. Finally, in light of the structural data now available, we attempt to interpret the likely consequences of mutations already identified in the gene encoding the h5beta-red enzyme which lead to a reduction of its enzymatic activity and which can progress to severe liver function failure.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/chemistry , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , Catalytic Domain , Ketosteroids/chemistry , Oxidoreductases/chemistry , Oxidoreductases/physiology , Binding Sites , Catalysis , Crystallography, X-Ray , Humans , Oxidation-Reduction
4.
Biochim Biophys Acta ; 1778(5): 1324-34, 2008 May.
Article in English | MEDLINE | ID: mdl-18284914

ABSTRACT

Lecithin retinol acyltransferase (LRAT) is a 230 amino acids membrane-associated protein which catalyzes the esterification of all-trans-retinol into all-trans-retinyl ester. The enzymatic activity of a truncated form of LRAT (tLRAT) which contains the residues required for catalysis but which is lacking N- and C-terminal hydrophobic segments has been shown to depend on the detergent used for its solubilization. Moreover, it is unknown whether tLRAT can bind membranes in the absence of these hydrophobic segments. The present study has allowed to measure the membrane binding and hydrolytic action of tLRAT in lipid monolayers by use of polarization modulation infrared reflection absorption spectroscopy and Brewster angle microscopy. Moreover, the proportion of the secondary structure components of tLRAT was determined in three different detergents by infrared absorption spectroscopy, vibrational circular dichroism and electronic circular dichroism which allowed to explain its detergent dependent activity. In addition, the secondary structure of tLRAT in the absence of detergent was very similar to that in Triton X-100 thus suggesting that, compared to the other detergents assayed, the secondary structure of this protein is very little perturbed by this detergent.


Subject(s)
Acyltransferases/chemistry , Acyltransferases/metabolism , Base Sequence , Circular Dichroism , DNA Primers , Detergents/chemistry , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrophotometry, Infrared
5.
J Biol Chem ; 282(42): 30910-9, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17711855

ABSTRACT

Antiandrogens are commonly used to treat androgen-dependent disorders. The currently used drugs unfortunately possess very weak affinity for the human AR (hAR), thus indicating the need to develop new high-affinity steroidal antiandrogens. Our compounds are specially designed to impede repositioning of the mobile carboxyl-terminal helix 12, which blocks the ligand-dependent transactivation function (AF-2) located in the AR ligand-binding domain (ARLBD). Using crystal structures of the hARLBD, we first found that H12 could be directly reached from the ligand-binding pocket (LBP) by a chain positioned on the C18 atom of an androgen steroid nucleus. A set of 5alpha-dihydrotestosterone-derived molecules bearing various C18 chains were thus synthesized and tested for their capacity to bind hAR and act as antagonists. Although most of those having very high affinity for hAR were agonists, several very potent antagonists were obtained, confirming the structural importance of the C18 chain. To understand the role of the C18 chain in their agonistic/antagonistic properties, the structure of the hARLBD complexed with one of these agonists, EM5744, was determined at a 1.65-A resolution. We have identified new interactions involving Gln(738), Met(742), and His(874) that explain both the high affinity of this compound and the inability of its bulky chain to prevent the repositioning of H12. This structural information will be helpful to refine the structure of the chains placed on the C18 atom to obtain efficient H12-directed steroidal antiandrogens.


Subject(s)
Androgen Antagonists/chemistry , Gestrinone/analogs & derivatives , Receptors, Androgen/chemistry , Androgen Antagonists/chemical synthesis , Androgen Antagonists/therapeutic use , Androgens , Crystallography, X-Ray , Dihydrotestosterone/antagonists & inhibitors , Dihydrotestosterone/chemistry , Gestrinone/chemical synthesis , Gestrinone/chemistry , Gonadal Disorders/drug therapy , Humans , Ligands , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary/physiology , Structure-Activity Relationship , Transcriptional Activation/drug effects
6.
J Mol Biol ; 369(2): 525-40, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17442338

ABSTRACT

The mouse 17alpha-hydroxysteroid dehydrogenase (m17alpha-HSD) is the unique known member of the aldo-keto reductase (AKR) superfamily able to catalyze efficiently and in a stereospecific manner the conversion of androstenedione (Delta4) into epi-testosterone (epi-T), the 17alpha-epimer of testosterone. Structural and mutagenic studies had already identified one of the residues delineating the steroid-binding cavity, A24, as the major molecular determinant for the stereospecificity of m17alpha-HSD. We report here a ternary complex crystal structure (m17alpha-HSD:NADP(+):epi-T) determined at 1.85 A resolution that confirms this and reveals a unique steroid-binding mode for an AKR enzyme. Indeed, in addition to the interactions found in all other AKRs (van der Waals contacts stabilizing the core of the steroid and the hydrogen bonds established at the catalytic site by the Y55 and H117 residues with the oxygen atom of the ketone group to be reduced), m17alpha-HSD establishes with the other extremity of the steroid nucleus an additional interaction involving K31. By combining direct mutagenesis and kinetic studies, we found that the elimination of this hydrogen bond did not affect the affinity of the enzyme for its steroid substrate but led to a slight but significant increase of its catalytic efficiency (k(cat)/K(m)), suggesting a role for K31 in the release of the steroidal product at the end of the reaction. This previously unobserved steroid-binding mode for an AKR is similar to that adopted by other steroid-binding proteins, the hydroxysteroid dehydrogenases of the short-chain dehydrogenases/reductases (SDR) family and the steroid hormone nuclear receptors. Mutagenesis and structural studies made on the human type 3 3alpha-HSD, a closely related enzyme that shares 73% amino acids identity with the m17alpha-HSD, also revealed that the residue at position 24 of these two enzymes directly affects the binding and/or the release of NADPH, in addition to its role in their 17alpha/17beta stereospecificity.


Subject(s)
Hydroxysteroid Dehydrogenases , Protein Structure, Tertiary , Steroids/metabolism , Animals , Binding Sites , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Humans , Hydroxysteroid Dehydrogenases/chemistry , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Mice , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , NADP/chemistry , NADP/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Steroids/chemistry
7.
Bioorg Med Chem ; 15(8): 3003-18, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17336533

ABSTRACT

In our efforts to develop compounds with therapeutic potential as antiandrogens, we synthesized a series of 5alpha-androstane-3alpha,17beta-diol derivatives with a fixed side-chain length of 3-methylenes at C-16alpha, but bearing a diversity of functional groups at the end. Among these, the chloride induced the best antiproliferative activity on androgen-sensitive Shionogi cells. Substituting the OH at C-3 by a methoxy group showed the importance of the OH. Moreover, its transformation into a ketone increased the androgen receptor (AR) binding but decreased the antiproliferative activity and induced a proliferative effect on Shionogi cells. These results confirm the importance of keeping a 5alpha-androstane-3alpha,17beta-diol nucleus instead of a dihydrotestosterone nucleus. Variable side-chain lengths of 2-, 3-, 4-, and 6-methylenes at C-16alpha were investigated and the optimal length was found to be 3-methylenes. Although exhibiting a weak AR binding affinity, 16alpha-(3'-chloropropyl)-5alpha-androstane-3alpha,17beta-diol (15) provided an antiproliferative activity on Shionogi cells similar to that of pure non-steroidal antiandrogen hydroxy-flutamide (77% and 67%, respectively, at 0.1 microM). The new steroidal compound, 15, thus constitutes a good starting point for development of future antiandrogens with a therapeutic potential against prostate cancer.


Subject(s)
Androgen Antagonists/chemical synthesis , Androgen Antagonists/pharmacology , Androstane-3,17-diol/chemical synthesis , Androstane-3,17-diol/pharmacology , Antineoplastic Agents, Hormonal/chemical synthesis , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Indicators and Reagents , Magnetic Resonance Spectroscopy , Models, Molecular , Receptors, Androgen/drug effects , Spectrophotometry, Infrared , Structure-Activity Relationship
8.
J Mol Biol ; 364(4): 747-63, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17034817

ABSTRACT

Very recently, the mouse 17alpha-hydroxysteroid dehydrogenase (m17alpha-HSD), a member of the aldo-keto reductase (AKR) superfamily, has been characterized and identified as the unique enzyme able to catalyze efficiently and in a stereospecific manner the conversion of androstenedione (Delta4) into epitestosterone (epi-T), the 17alpha-epimer of testosterone. Indeed, the other AKR enzymes that significantly reduce keto groups situated at position C17 of the steroid nucleus, the human type 3 3alpha-HSD (h3alpha-HSD3), the human and mouse type 5 17beta-HSD, and the rabbit 20alpha-HSD, produce only 17beta-hydroxy derivatives, although they possess more than 70% amino acid identity with m17alpha-HSD. Structural comparisons of these highly homologous enzymes thus offer an excellent opportunity of identifying the molecular determinants responsible for their 17alpha/17beta-stereospecificity. Here, we report the crystal structure of the m17alpha-HSD enzyme in its apo-form (1.9 A resolution) as well as those of two different forms of this enzyme in binary complex with NADP(H) (2.9 A and 1.35 A resolution). Interestingly, one of these binary complex structures could represent a conformational intermediate between the apoenzyme and the active binary complex. These structures provide a complete picture of the NADP(H)-enzyme interactions involving the flexible loop B, which can adopt two different conformations upon cofactor binding. Structural comparison with binary complexes of other AKR1C enzymes has also revealed particularities of the interaction between m17alpha-HSD and NADP(H), which explain why it has been possible to crystallize this enzyme in its apo form. Close inspection of the m17alpha-HSD steroid-binding cavity formed upon cofactor binding leads us to hypothesize that the residue at position 24 is of paramount importance for the stereospecificity of the reduction reaction. Mutagenic studies have showed that the m17alpha-HSD(A24Y) mutant exhibited a completely reversed stereospecificity, producing testosterone only from Delta4, whereas the h3alpha-HSD3(Y24A) mutant acquires the capacity to metabolize Delta4 into epi-T.


Subject(s)
Hydroxysteroid Dehydrogenases/chemistry , Animals , Apoenzymes , Binding Sites , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Mice , Models, Molecular , Mutagenesis, Site-Directed , NADP , Protein Binding , Stereoisomerism
9.
Protein Sci ; 15(5): 987-99, 2006 May.
Article in English | MEDLINE | ID: mdl-16641486

ABSTRACT

Androgens exert their effects by binding to the highly specific androgen receptor (AR). In addition to natural potent androgens, AR binds a variety of synthetic agonist or antagonist molecules with different affinities. To identify molecular determinants responsible for this selectivity, we have determined the crystal structure of the human androgen receptor ligand-binding domain (hARLBD) in complex with two natural androgens, testosterone (Testo) and dihydrotestosterone (DHT), and with an androgenic steroid used in sport doping, tetrahydrogestrinone (THG), at 1.64, 1.90, and 1.75 A resolution, respectively. Comparison of these structures first highlights the flexibility of several residues buried in the ligand-binding pocket that can accommodate a variety of ligand structures. As expected, the ligand structure itself (dimension, presence, and position of unsaturated bonds that influence the geometry of the steroidal nucleus or the electronic properties of the neighboring atoms, etc.) determines the number of interactions it can make with the hARLBD. Indeed, THG--which possesses the highest affinity--establishes more van der Waals contacts with the receptor than the other steroids, whereas the geometry of the atoms forming electrostatic interactions at both extremities of the steroid nucleus seems mainly responsible for the higher affinity measured experimentally for DHT over Testo. Moreover, estimation of the ligand-receptor interaction energy through modeling confirms that even minor modifications in ligand structure have a great impact on the strength of these interactions. Our crystallographic data combined with those obtained by modeling will be helpful in the design of novel molecules with stronger affinity for the AR.


Subject(s)
Androgen Antagonists/chemistry , Multiprotein Complexes , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Androgen Antagonists/metabolism , Androgens , Binding Sites , Crystallization , Crystallography, X-Ray , Dihydrotestosterone/chemistry , Dihydrotestosterone/metabolism , Gestrinone/analogs & derivatives , Gestrinone/chemistry , Gestrinone/metabolism , Humans , Hydrogen Bonding , Ligands , Molecular Structure , Protein Binding , Protein Structure, Tertiary , Testosterone/chemistry , Testosterone/metabolism
10.
Biochemistry ; 45(10): 3337-47, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16519528

ABSTRACT

RPE65 is the major component of the retinal pigment epithelium (RPE) microsomal membrane, and it plays a critical role in the binding of retinoids involved in the visual cycle. To understand how RPE65 binds to membranes, we have expressed and purified soluble fragments of human RPE65 fused to glutathione S-transferase (GST). The interaction between two fragments of RPE65 (F1 and F2 which include residues 1-125 and 126-250, respectively) and lipid monolayers has been studied by surface pressure, ellipsometry, and surface rheology measurements. Surface pressure and ellipsometry clearly showed a rapid adsorption of F2 to lipid monolayers whereas the kinetics of binding of F1 was much slower. Furthermore, the data suggest that the F2 fragment inserts into the lipid monolayer. Surface rheology showed a clear increase in monolayer rigidity only in the presence of F2, thereby demonstrating high intermolecular interactions of this fragment. This observation is further supported by the GST pull-down assays which demonstrated that F2 cosediments with full-length RPE65, suggesting that RPE65 has the propensity to form clusters or oligomers. The structure homology modeling of RPE65 based on a related family member, apocarotene 15',15'-oxygenase, further suggests that a hydrophobic patch located in the F2 region might be responsible for membrane binding. The present work shows that F2 interacts much stronger with lipid monolayers than does F1, which suggests that the region of RPE65 located between residues 126-250 should be very important for its membrane binding. Moreover, given that these fragments are not acylated, these data also suggest that an effective binding of RPE65 to membranes can be achieved without palmitoylation. Furthermore, GST pull-down assays also indicated that F2 interacts with 11-cis-retinol dehydrogenase, which supports previous data suggesting that it could act as a partner of RPE65.


Subject(s)
Cell Membrane/metabolism , Eye Proteins/metabolism , Glutathione Transferase/metabolism , Amino Acid Sequence , Animals , Baculoviridae/metabolism , Carrier Proteins , Escherichia coli/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Insecta/metabolism , Molecular Sequence Data , Oxidoreductases/metabolism , Oxygenases/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solubility , Time Factors , beta-Carotene 15,15'-Monooxygenase/metabolism , cis-trans-Isomerases
11.
BMC Biochem ; 6: 12, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-16018803

ABSTRACT

BACKGROUND: Epi-testosterone (epiT) is the 17alpha-epimer of testosterone. It has been found at similar level as testosterone in human biological fluids. This steroid has thus been used as a natural internal standard for assessing testosterone abuse in sports. EpiT has been also shown to accumulate in mammary cyst fluid and in human prostate. It was found to possess antiandrogenic activity as well as neuroprotective effects. So far, the exact pathway leading to the formation of epiT has not been elucidated. RESULTS: In this report, we describe the isolation and characterization of the enzyme 17alpha-hydroxysteroid dehydrogenase. The name is given according to its most potent activity. Using cells stably expressing the enzyme, we show that 17alpha-HSD catalyzes efficienty the transformation of 4-androstenedione (4-dione), dehydroepiandrosterone (DHEA), 5alpha-androstane-3,17-dione (5alpha-dione) and androsterone (ADT) into their corresponding 17alpha-hydroxy-steroids : epiT, 5-androstene-3beta,17alpha-diol (epi5diol), 5alpha-androstane-17alpha-ol-3-one (epiDHT) and 5alpha-androstane-3alpha,17alpha-diol (epi3alpha-diol), respectively. Similar to other members of the aldo-keto reductase family that possess the ability to reduce the keto-group into hydroxyl-group at different position on the steroid nucleus, 17alpha-HSD could also catalyze the transformation of DHT, 5alpha-dione, and 5alpha-pregnane-3,20-dione (DHP) into 3alpha-diol, ADT and 5alpha-pregnane-3alpha-ol-20-one (allopregnanolone) through its less potent 3alpha-HSD activity. We also have over-expressed the 17alpha-HSD in Escherichia coli and have purified it by affinity chromatography. The purified enzyme exhibits the same catalytic properties that have been observed with cultured HEK-293 stably transfected cells. Using quantitative Realtime-PCR to study tissue distribution of this enzyme in the mouse, we observed that it is expressed at very high levels in the kidney. CONCLUSION: The present study permits to clarify the biosynthesis pathway of epiT. It also offers the opportunity to study gene regulation and function of this enzyme. Further study in human will allow a better comprehension about the use of epiT in drug abuse testing; it will also help to clarify the importance of its accumulation in breast cyst fluid and prostate, as well as its potential role as natural antiandrogen.


Subject(s)
Epitestosterone/biosynthesis , Hydroxysteroid Dehydrogenases/chemistry , Hydroxysteroid Dehydrogenases/metabolism , Amino Acid Sequence , Animals , Cell Line , Enzyme Activation/physiology , Epitestosterone/genetics , Female , Humans , Hydroxysteroid Dehydrogenases/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data
12.
Protein Sci ; 14(6): 1485-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15929998

ABSTRACT

The aldo-keto reductase (AKR) human type 3 3alpha-hydroxysteroid dehydrogenase (h3alpha-HSD3, AKR1C2) plays a crucial role in the regulation of the intracellular concentrations of testosterone and 5alpha-dihydrotestosterone (5alpha-DHT), two steroids directly linked to the etiology and the progression of many prostate diseases and cancer. This enzyme also binds many structurally different molecules such as 4-hydroxynonenal, polycyclic aromatic hydrocarbons, and indanone. To understand the mechanism underlying the plasticity of its substrate-binding site, we solved the binary complex structure of h3alpha-HSD3-NADP(H) at 1.9 A resolution. During the refinement process, we found acetate and citrate molecules deeply engulfed in the steroid-binding cavity. Superimposition of this structure with the h3alpha-HSD3-NADP(H)-testosterone/acetate ternary complex structure reveals that one of the mobile loops forming the binding cavity operates a slight contraction movement against the citrate molecule while the side chains of many residues undergo numerous conformational changes, probably to create an optimal binding site for the citrate. These structural changes, which altogether cause a reduction of the substrate-binding cavity volume (from 776 A(3) in the presence of testosterone/acetate to 704 A(3) in the acetate/citrate complex), are reminiscent of the "induced-fit" mechanism previously proposed for the aldose reductase, another member of the AKR superfamily. We also found that the replacement of residues Arg(301) and Arg(304), localized near the steroid-binding cavity, significantly affects the 3alpha-HSD activity of this enzyme toward 5alpha-DHT and completely abolishes its 17beta-HSD activity on 4-dione. All these results have thus been used to reevaluate the binding mode of this enzyme for androgens.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/chemistry , Androgens/chemistry , 3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/metabolism , Amino Acid Motifs , Androgens/metabolism , Crystallography, X-Ray , Humans , Protein Binding , Protein Structure, Tertiary
13.
Biochim Biophys Acta ; 1679(3): 230-47, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15358515

ABSTRACT

The human ELAC2 gene was the first candidate prostate cancer susceptibility gene identified by linkage analysis and positional cloning. DNA sequence indicates a protein of 826 amino acids encoded by 24 exons. In the present study, we characterized the coding sequence of chimpanzee and gorilla ELAC2 orthologs by direct sequencing of genomic fragments, and of cynomolgus monkey and rat orthologs by screening cDNA libraries. The orthologs characterized in the chimpanzee, gorilla and cynomolgus monkey also encode proteins of 826 amino acids, sharing 98.9%, 98.5% and 93.7% sequence identity with the human protein. Our analyses of the mouse ELAC2 gene identified two alternative mRNA transcripts. One is translated into a protein of 824 a.a. (mouse ELAC2), whereas the other one encodes a protein of 831 amino acids (mouse ELAC2A) resulting from an alternatively spliced form of 25 exons. The rat ELAC2 gene ortholog also expressed two similar alternatively spliced transcripts. These two forms are ubiquitously expressed in mouse and rat tissues. The highest levels of expression of the ELAC2 form are observed in the testis while the lowest levels are seen in the prostate and in the muscle. However, it is of interest to note that the relative abundance of the rat and mouse ELAC2 transcripts, measured by real-time quantitative PCR, is higher than the respective ELAC2A forms in all surveyed tissues except for the prostate and the muscle. The ELAC2A transcript levels are 4.1 to 5.0-fold higher than the ELAC2 levels in the prostate of rat and mouse, respectively. A fine analysis of the conserved domains on the primary structure of ELAC2 orthologs revealed the presence of a putative beta-CASP domain shared by the PSO2 (SNM1) DNA interstrand cross-link repair proteins, and the 73-kDa subunit of mRNA 3' end cleavage and polyadenylation specificity factor (CPSF73) as well as Artemis proteins, thus suggesting a potential interaction of ELAC2 gene product with nucleic acids and more specifically with RNA targets. Taken together, these data offer useful tools to further study the regulation and cellular function of ELAC2 gene in experimental models and provide further insight concerning conserved amino acid motifs that could have biological significance.


Subject(s)
Neoplasm Proteins/genetics , Primates/genetics , Prostatic Neoplasms/genetics , Rodentia/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cleavage And Polyadenylation Specificity Factor/metabolism , Cloning, Molecular , Conserved Sequence , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Male , Mice , Molecular Sequence Data , Mutation, Missense , Neoplasm Proteins/metabolism , Organ Specificity , Protein Structure, Tertiary , Rats , Testis/physiology
14.
J Mol Biol ; 339(1): 89-102, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15123423

ABSTRACT

The aldo-keto reductase rabbit 20alpha-hydroxysteroid dehydrogenase (rb20alpha-HSD; AKR1C5) is less selective than other HSDs, since it exerts its activity both on androgens (C19 steroids) and progestins (C21 steroids). In order to identify the molecular determinants responsible for this reduced selectivity, binary (NADPH) and ternary (NADP(+)/testosterone) complex structures were solved to 1.32A and 2.08A resolution, respectively. Inspection of the cofactor-binding cavity led to the identification of a new interaction between side-chains of residues His222 and Lys270, which cover the central phosphate chain of the cofactor, reminiscent of the "safety-belt" found in other aldo-keto reductases. Testosterone is stabilized by a phenol/benzene tunnel composed of side-chains of numerous residues, among which Phe54, which forces the steroid to take up an orientation markedly contrasting with that found in HSD ternary complexes reported. Combining structural, site-directed mutagenesis, kinetic and fluorescence titration studies, we found that the selectivity of rb20alpha-HSD is mediated by (i) the relaxation of loop B (residues 223-230), partly controlled by the nature of residue 230, (ii) the nature of the residue found at position 54, and (iii) the residues found in the C-terminal tail of the protein especially the side-chain of the amino acid 306.


Subject(s)
20-alpha-Hydroxysteroid Dehydrogenase/chemistry , 20-alpha-Hydroxysteroid Dehydrogenase/metabolism , NADP/metabolism , Testosterone/metabolism , 20-alpha-Hydroxysteroid Dehydrogenase/genetics , Animals , Arginine/metabolism , Binding Sites , Crystallization , Escherichia coli/enzymology , Fluorescence , Glutathione Transferase/metabolism , Histidine/metabolism , Hydrogen Bonding , Kinetics , Mutagenesis, Site-Directed , Protein Conformation , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Steroids/chemistry , Steroids/metabolism , Substrate Specificity , Testosterone/chemistry
15.
Biochem Biophys Res Commun ; 310(3): 767-73, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14550269

ABSTRACT

The recently discovered CLECSF6 protein displays the features of a receptor involved in the down-modulation of leukocyte activation. Although CLECSF6 has been the focus of the interest of many researchers lately, a Western blot characterization of the protein is still lacking. This highly reduces our ability to gain full knowledge of the biological relevance of this protein in cell responses. We produced two rabbit polyclonal antisera that detected a glycosylated protein at approximately 35kDa in neutrophils. Four different CLECSF6 mRNA species have been discovered to date. When deglycosylated, the protein displayed the molecular weight expected for the longest CLECSF6 form. Neutrophil membrane fractions were strongly enriched in the protein. We showed a down-modulation of the expression of this protein in neutrophils treated with granulocyte-macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor (TNF-alpha), lipopolysaccharide (LPS), and interleukin (IL)-4. This work supports the hypothesis that CLECSF6 is involved in the control of inflammation in neutrophils.


Subject(s)
Down-Regulation , Lectins, C-Type/biosynthesis , Membrane Glycoproteins , Neutrophils/metabolism , Receptors, Immunologic/biosynthesis , Animals , Baculoviridae/metabolism , Blotting, Western , Cell Line , Cell Membrane/metabolism , Cytokines/metabolism , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Glycosylation , Humans , Immunoblotting , Insecta , Interleukin-4/metabolism , Leukocytes/metabolism , Lipopolysaccharides/metabolism , Peptides/chemistry , RNA, Messenger/metabolism , Rabbits , Recombinant Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
J Mol Biol ; 331(3): 593-604, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12899831

ABSTRACT

Human 20alpha-hydroxysteroid dehydrogenase (h20alpha-HSD; AKR1C1) catalyzes the transformation of progesterone (Prog) into 20alpha-hydroxy-progesterone (20alpha-OHProg). Although h20alpha-HSD shares 98% sequence identity with human type 3 3alpha-HSD (h3alpha-HSD3, AKR1C2), these two enzymes differ greatly in their activities. In order to explain these differences, we have solved the crystal structure of h20alpha-HSD in a ternary complex with NADP(+) and 20alpha-OHProg at 1.59A resolution. The steroid is stabilized by numerous hydrophobic interactions and a hydrogen bond between its O20 and the N(epsilon ) atom of His222. This new interaction prevents the formation of a hydrogen bond with the cofactor, as seen in h3alpha-HSD3 ternary complexes. By combining structural, direct mutagenesis and kinetic studies, we found that the H(222)I substitution decreases the K(m) value for the cofactor 95-fold. With these results, we hypothesize that the rotation of the lateral chain of His222 could be a mediating step between the transformation of Prog and the release of the cofactor. Moreover, crystal structure analysis and direct mutagenesis experiments lead us to identify a new residue involved in the binding of Prog. Indeed, the R(304)L substitution leads to a 65-fold decrease in the K(m) value for Prog reduction. We thus propose that Prog is maintained in a new steroid-binding site composed mainly of residues found in the carboxy-terminal region of the protein.


Subject(s)
20-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/metabolism , Steroids/metabolism , 20-Hydroxysteroid Dehydrogenases/genetics , 20-alpha-Hydroxysteroid Dehydrogenase , Arginine/metabolism , Binding Sites , Crystallization , Histidine/metabolism , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Kinetics , Leucine/metabolism , Ligands , Models, Molecular , Mutagenesis, Site-Directed , NADP/metabolism , Progesterone/analogs & derivatives , Progesterone/chemistry , Progesterone/metabolism , Protein Conformation , Rotation , Stereoisomerism , Steroids/chemistry , Substrate Specificity , X-Ray Diffraction
17.
Acta Crystallogr D Biol Crystallogr ; 58(Pt 1): 135-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752791

ABSTRACT

Progesterone plays an essential role in the maintenance of the pregnancy of most mammals. 20alpha-Hydroxysteroid dehydrogenase (20alpha-HSD) catalyses the inactivation of progesterone into its inactive form, 20alpha-hydroxyprogesterone, and could thus be involved in progesterone withdrawal and in the control of gestation. In this report, the purification and crystallization of recombinant human and rabbit 20alpha-HSDs (h20alpha-HSD and rb20alpha-HSD) are described, two highly homologous enzymes possessing, in addition to their common 20alpha-HSD activity, different activities and substrate specificities. Complete diffraction data sets have been collected for crystals of rb20alpha-HSD in complex with NADP(H) and with either dihydrotestosterone (1.8 A), progesterone (1.7 A) or 4-androstenedione (1.8 A). All these crystals belong to the monoclinic space group P2(1). A partial data set has also been collected for a crystal of h20alpha-HSD (P2(1)2(1)2(1)) in complex with NADP(H) and progesterone.


Subject(s)
20-Hydroxysteroid Dehydrogenases/chemistry , NADP/metabolism , Steroids/metabolism , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Humans , Molecular Sequence Data , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...