227 REVIEW

Unique multifunctional HSD17B4 product: 17â-hydroxysteroid dehydrogenase 4 and D-3-hydroxyacyl- coenzyme A dehydrogenase/hydratase involved in Zellweger syndrome

Y de Launoit1 and J Adamski2 1Virology Unit, Faculty of Medicine, CP 614, Free University Brussels, 808 route de Lennik, 1070 Brussels, Belgium 2GSF National Research Center for Environment and Health, Institute of Mammalian Genetics, Genome Analysis Center, 85764 Neuherberg, Germany (Requests for offprints should be addressed to Y de Launoit)

ABSTRACT Six types of human 17â-hydroxysteroid dehydro- able to perform the dehydrogenase reaction not only genases catalyzing the conversion of estrogens and with at the C17 position but also with androgens at position C17 have been identified so -3-hydroxyacyl-coenzyme A (CoA). The enzyme far. The peroxisomal 17â-hydroxysteroid dehydro- is not active with -stereoisomers. The central part genase type 4 (17â-HSD 4, gene name HSD17B4) of the 80 kDa (amino acids 324–596) catalyzes the oxidation of estradiol with high catalyzes the 2-enoyl-acyl-CoA hydratase reaction preference over the reduction of . The with high efficiency. The C-terminal part of the highest levels of 17â-HSD 4 mRNA transcription 80 kDa protein (amino acids 597–737) facilitates the and specific activity are found in liver and kidney transfer of 7-dehydrocholesterol and phosphatidyl- followed by ovary and testes. A 3 kb mRNA codes choline between membranes in vitro. The for an 80 kDa (737 amino acids) protein featuring HSD17B4 gene is stimulated by progesterone, and domains which are not present in the other ligands of PPARá (peroxisomal proliferator acti- 17â-HSDs. The N-terminal domain of 17â-HSD 4 vated receptor alpha) such as clofibrate, and is reveals only 25% amino acid similarity with the down-regulated by phorbol esters. Mutations in the other types of 17â-HSDs. The 80 kDa protein is HSD17B4 lead to a fatal form of Zellweger N-terminally cleaved to a 32 kDa enzymatically syndrome. active fragment. Both the 80 kDa and the Journal of Molecular Endocrinology (1999) 22, 227–240 N-terminal 32 kDa (amino acids 1–323) protein are

INTRODUCTION names are used frequently. The complexity of related enzymes was reviewed recently elsewhere The genetics and biochemistry of conversion (Reed 1991, Andersson 1995, Poutanen et al. 1995, have recently been developing very fast. There is a Penning 1997). growing body of reports showing the involvement of steroid converting enzymes in several human disorders. In this review we will focus on the most MULTIPLE DEHYDROGENASES CONVERT unique protein among the 17â-hydroxysteroid STEROIDS AT POSITION C17 dehydrogenases, the type 4 enzyme. For the sake of clarity we will use the abbreviation 17â-HSD 4, Biological potency of estrogens and androgens is although, as will be discussed later, alternative regulated by conversions at position C17 by

Journal of Molecular Endocrinology (1999) 22, 227–240 Online version via http://www.endocrinology.org 0952–5041/99/022–227  1999 Society for Endocrinology Printed in Great Britain Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 228    and   · 17â-HSD 4

 1. Human 17â-hydroxysteroid dehydrogenases

Subcellular Mass Km nmol/mg Catalysis Tissue localization (kDa) Best substrate (ìM) protein in vivo Enzyme 17â-HSD 1 Placenta Soluble 34 Estrone 8·6 0·02 Reduction (Peltoketo et al. 1988) Ovary Estradiol 5·9 17â-HSD 2 Placenta Microsomal 43 Testosterone 0·4 0·22 Oxidation (Wu et al. 1993) Liver Estradiol 0·2 17â-HSD 3 Testis Microsomal 35 Androstenedione 0·5 0·09 Reduction (Geissler et al. 1994) Estrone 0·5 17â-HSD 4 Liver Peroxisomal 80 Estradiol 0·2 0·14 Oxidation (Adamski et al. 1995) Kidney Ä5-Androstenediol 0·4 17â-HSD 5 Liver Microsomal 34 5á-Dihydrotestosterone 19·0 2·45 Reduction (Lin et al. 1997; Zhang et al. 1995) Testis 3á-Androstanediol 17·1 4·83 Oxidation

17â-hydroxysteroid dehydrogenases (17â-HSDs). et al. 1997). Later, we will discuss the issue of Several enzymes with close substrate specificity multifunctionality in more detail. participate in that process. The identification and The oxidative 17â-HSD activity found in human characterization of individual human 17â-HSDs uterus endometrium could not be unequivocally was limited by the minute amounts of tissue ascribed to the known enzymes (Tseng & Mazella available for purification. However, analyses per- 1981). Attempts to isolate the endometrial 17â- formed with homogenates or with subcellular HSD from the particulate fraction of homogenates fractions allowed the kinetical differentiation of (Pollow et al. 1976) resulted in a 40-fold enrich- several enzymes such as the soluble 17â- ment. However, because of difficulties in collecting hydroxysteroid oxidoreductase of placenta or the and the paucity of starting material, the enriched structure-associated 17â-estradiol dehydrogenase fractions were not applied to amino acid sequencing of uterus epithelium (Engel & Groman 1974, Tseng and antibody production. Entenmann et al. (1980) & Gurpide 1974, Tseng & Mazella 1981). Before discovered oxidative activity for 17â-estradiol in molecular biology techniques became widespread, porcine endometrium. This microsomal activity the readily available human placenta allowed the revealed comparable kinetical parameters (NAD+- purification of the first human 34 kDa 17â- dependency, Km less than 1 µM). The parameters hydroxysteroid dehydrogenase (17â-HSD 1), which suggested a role in the inactivation of hormones. became a model for studies of steroid converting enzymes. After cloning and elucidation of the gene structure (Peltoketo et al. 1988, Luu-The et al. PURIFICATION OF PORCINE 17â-ESTRADIOL 1989), it represents the best characterized human DEHYDROGENASE 4 steroid dehydrogenase. In vivo this enzyme participates in the synthesis of steroids via a To accomplish identification of a novel 17â-HSD reductive pathway. However, detailed kinetical we have used a pig (Sus scrofa) model. The studies (Blomquist et al. 1985) have shown that the epithelial layer of the porcine uterus could easily placenta expresses additional HSDs and one of be collected at a preparative scale by curettage them, namely 17â-HSD 2, was cloned (Wu et al. (Sierralta et al. 1978). The 17â-estradiol dehydro- 1993) (Table 1). This enzyme is a 43 kDa genase activity had to be solubilized from the microsomal dehydrogenase revealing a twofold particulate fraction (Adamski et al. 1992a). Extracts higher rate of oxidation than reduction for both were applied to DEAE-Sepharose, depleted of free estrogens and androgens. A further enzyme, detergent on Amberlite XAD-2 and further purified 17â-HSD 3, is most abundant in testes and by affinity chromatography on blue Sepharose represents a transmembrane microsomal 35 kDa (Adamski et al. 1992b). Further purification on protein with a strong preference for the reduction of butyl Sepharose resulted in two products rich in androgens (Geissler et al. 1994). Mouse and human 17â-estradiol dehydrogenase activity: a major mod- 17â-HSD type 5 has not yet been fully character- erately hydrophobic fraction (EDH) and a minor ized (Deyashiki et al. 1995, Zhang et al. 1995). very hydrophobic fraction (VHF). They were However, this enzyme has wide tissue distri- processed in parallel by gel filtration and ion- bution and has also been identified as 3á-HSD type exchange chromatography on Mono S. The EDH 2 (Lin et al. 1997) (for a review see Penning fraction was purified to homogeneity and revealed a

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   229

 2. Kinetic parameters of purified porcine this fragment as a probe, a 3 kb cDNA was isolated 17â-estradiol dehydrogenase 4 from a porcine ëZAP kidney cDNA library. The sequence was later confirmed in porcine uterus E E E E 2< 1 2= 1 (Leenders et al. 1994a). The cDNA coded for a Parameter protein of 80 kDa consisting of 737 aa and was not Optimal pH 7·8 6·6 similar to any known steroid dehydrogenases. Km for steroid 0·22 ìM 1·10 mM Best cofactor NAD+ NADPH About 70% of its amino acid sequence was already known from peptides of the 32 and 80 kDa . Km for cofactor 44 mM 21 mM Screening of human and mouse cDNA ëgt11

E2,17â-estradiol; E1, estrone. libraries of liver was performed with porcine enzyme cDNA. Novel 3 kb cDNAs were identified which coded for proteins of 735 and 736 aa single band at 32 kDa in the denaturing SDS- representing the human and mouse counterparts PAGE. The VHF was a mixture of proteins of 32, respectively of the porcine enzyme (Adamski et al. 45 and 80 kDa (Adamski et al. 1992b). 1995, Normand et al. 1995).

CHARACTERIZATION OF PURIFIED PARALLEL WAYS OF HSD17B4 PORCINE 17â-HSD 4 IDENTIFICATION

Both purification products, the EDH and the VHF, The product of the HSD17B4 gene, an 80 kDa show the same Km for steroids and cofactors as protein, was detected almost in parallel by other those measured with two-substrate kinetics in the groups. During studies on peroxisomal â-oxidation particulate fraction of homogenates of porcine of pristanic acid and bile acid intermediates in rat uterus epithelium (Table 2). They reveal an ordered and man, 80 kDa -specific hydroxyacyl-coenzyme mechanism of reaction with the cofactor binding A (CoA) dehydrogenase/hydratase (also called first (Adamski et al. 1992b, Marks 1992). EDH and multifunctional protein 2–MFP2) were purified VHF also share the same substrate specificity, (Novikov et al. 1994, Qin et al. 1997b), character- which is highest for 17â-estradiol and, unexpect- ized and cloned (Dieuaide-Noubhani et al. 1996a, edly, comparably high for 5-androstene-3â,17â-diol 1997a,b, Jiang et al. 1996, 1997, Novikov et al. (Km=0·2 µM). Other androgens or progestagens are 1997). The amino acid sequence of the human not converted. enzyme was identical to that of 17â-HSD 4 The molecular mass was estimated in denaturing (Adamski et al. 1995). SDS-PAGE and under native conditions by gel Guinea pig enzyme was cloned from a cDNA filtration and density gradient centrifugation library while using rat 17â-HSD 4 as a probe (Caira (Adamski et al. 1992b). The 32 kDa protein has a et al. 1998). capability of forming dimers with an apparent Another study pursued the identification of molecular weight of 75 kDa (Carstensen et al. 1996). cDNAs up-regulated in rat by peroxisomal prolif- The interactions in the VHF are more complex. erators such as WY14,643 (Corton et al. 1996, Both gel filtration analyses and density gradient 1997). One of the affected proteins turned out to be centrifugation revealed the presence of a hetero- the rat ortholog of previously known porcine, mouse geneous complex with at least two molecular mass and human 17â-HSD 4. forms of 170 kDa and 240 kDa. Unusual was the way that chicken protein was identified (Kobayashi et al. 1997). A monoclonal antibody 3b5 was prepared against isolated retinal CLONING OF THE PORCINE, HUMAN AND pigment epithelium cells. The antibody recognized MOUSE 17â-ESTRADIOL DEHYDROGENASE a 75 kDa protein on western blots of retinal pigment TYPE 4 epithelium. This antibody was then used to screen a lambda expression library. This approach resulted With degenerated PCR-primers, designed accord- in the identification of a full length cDNA clone ing to partial amino acid (aa) sequence of the 32 kDa coding for the chicken 17â-HSD 4. protein, a fragment of 405 base pairs (bp) was Amino acid identities between 17â-HSD 4 from amplified from porcine endometrium cDNA. It had different species are very high, around 80% (Fig. 1). a single open reading frame coding for an amino Several hydroxysteroid dehydrogenases could be acid sequence which was identical to the 32 kDa traced to a common ancestor close to 3-ketoacyl- protein as confirmed by Edman degradation. Using acyl carrier protein reductase of Eschericia coli

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 230    and   · 17â-HSD 4

 1. Alignment of amino acid sequences of 17â-HSD 4 from different species. Sequences were ordered to obtain maximum similarity with ClustalX software. Identical amino acids are grey shaded, similar amino acids are boxed. Accession numbers for protein sequences are: mouse, P51660; chicken, U77911; rat, U37486; human, P51659; porcine, X78201; guinea pig, Y13623.

(Baker 1996). Phylogenetic studies suggest a GENE STRUCTURE common ancestor for the 17â-HSD 1 and 2, whereas the 17â-HSD 3 and 4 are examples of The chromosomal assignment and structure of the convergence (Baker 1994, 1996). In particular human HSD17B4 gene were determined recently 17â-HSD 4 appears to be a very ancient protein (Leenders et al. 1996a, Novikov et al. 1997, whose functionality and amino acid sequence are Leenders et al. 1999). The HSD17B4 gene localizes strongly conserved. Similarities to proteins of to 5q2 and was found to be more than primitive organisms will be discussed in the gene 100 kbp in length (Fig. 2). The size of the exons structure section. ranges from 21 bp (exon 5) to 286 bp (exon 13). The smallest intron with 100 bp was found between exons 5 and 6. NAMES USED The N-terminal domain of 17â-HSD 4 reveals functional similarity and amino acid homology Due to multifunctionality of the encoded protein, (54%) to the family of short chain alcohol the HSD17B4 gene product was discovered by dehydrogenases (SCAD) (Persson et al. 1991, many groups using different strategies. Because of Leenders et al. 1994b,Jo¨rnvall et al. 1995) and the that there are several names being used. Table 3 central domain to several fatty acid hydratases gives a summary of them. The references given (HDE), especially to that of Candida tropicalis (40%) should be understood as a source for the represen- (Baker 1996). However, neither sizes of the tative description. Quite often different synonyms corresponding nor the exon/intron structures are used in the same reports, depending on the are analogous. As discussed later, the 17â-HSD 4 is functionality analyzed. an exception in the group of 17â-HSDs, since it

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   231

 3. Names used for the HSD17B4 gene product

Reference Name 17â-hydroxysteroid dehydrogenase type 4 or 17â-HSD 4 Leenders et al. (1994) Multifunctional protein 2 or MFP-2 Dieuaide-Noubhani et al. (1996) Peroxisomal multifunctional enzyme 2 or perMFE2 Qin et al. (1997) -specific multifunctional protein 2 Caira et al. (1998) -3-hydroxyacyl-CoA dehydrogenase Dieuaide-Noubhani et al. (1997); Novikov et al. (1994) 2-trans-enoyl-CoA hydratase Dieuaide-Noubhani et al. (1997) -bifunctional protein Jiang et al. (1997) -3-hydroxyacyl-CoA dehydratase/ Jiang et al. (1996) -3-hydroxyacyl dehydrogenase bifunctional protein

 2. Structure of 17â-HSD 4 protein and HSD17B4 gene. A schematic structure of the human 17â-HSD IV protein is given above that of the gene. Arrows point to the position of the G16S mutation, cleavage site and C-terminal peroxisomal targeting signal. The black boxes representing exons are drawn to scale according to the indicated 10 kb marker. The sizes of introns are given above: introns smaller than 7 kb are drawn to scale, larger introns are represented by broken lines. The numbers of the exons are below the boxes. Exons 1 and 24 consist of translated (black box) and untranslated (grey box) regions. Accession numbers for the nucleotide sequence are AF057720-AF057740. consists of 3 domains. Only the N-terminal domain functionality as the C-terminal domain (sterol of 320 amino acids participates in steroid metab- carrier protein (SCP) 2) of the SCPX protein (Ohba olism and is coded by 12 exons spanning about et al. 1994, Leenders et al. 1996b). Interestingly, the 40 kbp. In comparison to other 17â-hydroxysteroid gene structure of the last 3 exons is also similar. dehydrogenases, the SCAD domain of the This observation supports the hypothesis that the HSD17B4 gene is much bigger than the 17â-HSD 1 HSD17B4 gene is the result of a gene fusion. (6 exons on 3·3 kbp) (Luu-The et al. 1990), close to the 17â-HSD 2 (7 exons on 40 kbp) (Labrie et al. 1995) and smaller than the 17â-HSD 3 (11 exons on PROCESSING OF 80 kDa PROTEIN 60 kbp) (Geissler et al. 1994). The amino acid sequence C-terminal domain of Some peroxisomal proteins (SCP2, 3-ketoacyl-CoA 17â-HSD 4 reveals 40% identity and the same ) are cleaved from larger precursors in the

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 232    and   · 17â-HSD 4

 4. Distribution of 17â-estradiol dehydrogenase activity in porcine tissues

E2

E2,17â-estradiol; E1, estrone; ND, not detected.

course of translocation (Swinkels et al. 1991). A Highest activities are found in liver and kidney protease present in has been proposed followed by uterus, lung, ovary and testes (Table 4). to recognize the sequence Ala-[AlaVal]-Pro (Mori Immunohistochemical analysis of tissues showing et al. 1991). The 80 kDa full length HSD17B4 gene low 17â-HSD 4 mRNA expression, such as brain, product is N-terminally cleaved, probably after lung and uterus, reveals that the enzyme is present the sequence Ala320-Ala-Pro-Ser324,toa32kDa in specific cells within these organs. In the rat and fragment representing a SCAD domain (Leenders mouse cerebellum 17â-HSD 4 is confined to et al. 1994b). Such processing has also been Purkinje cells, and the expression is also seen in the observed in mice and rats (Novikov et al. 1994, anterior pituitary (Normand et al. 1995). There is Normand et al. 1995, Dieuaide-Noubhani et al. high expression of 17â-HSD 4 in chick retinal 1997b) although the protease recognition motif was pigment epithelium but not in other parts of the eye not conserved. The meaning of this cleavage for (Kobayashi et al. 1997). In the lung the bronchial 17â-HSD 4 is not known. Interestingly, the extent epithelium expresses high levels of 17â-HSD 4 of processing of 80 kDa into 32 kDa varies among (Mo¨ller et al. 1999) and in the uterus the protein is porcine tissues (Adamski et al. 1997). Western blot present in luminal and glandular epithelium and not analyses of porcine organs revealed that target in stromal cells (Husen et al. 1994). tissues (uterus and mammary glands) show high A slightly different expression pattern was seen in processing. In these tissues the 32 kDa form human tissues. The highest mRNA level of human dominates. On the other hand, non-target tissues 17â-HSD 4 was observed in liver, followed by participating in â-oxidation of fatty acids (such as heart, prostate and testis. Moderate expression liver or kidney) showed low processing. Neverthe- occurred in lung, skeletal muscle, kidney, pancreas, less, in either tissue both proteins are present. The thymus, ovary, intestine and term placenta. Weak differential processing raised the question whether signals were observed in brain, spleen, colon and the release of the 32 kDa fragment from the 80 kDa lymphocytes. In all cases only a single band at 3 kb protein is an activation step for 17â-hydroxysteroid was detectable (Adamski et al. 1995). The wide dehydrogenase. However, both 80 and 32 kDa distribution of 17â-HSD 4 may in part explain proteins had comparable kinetical parameters after oxidative activities measured in human tissues transient expression in HEK 293 cells or puri- (Martel et al. 1992). The expression of 17â-HSD 4 fication of recombinant proteins from E. coli is in contrast with that of 17â-HSD 1 and 2 which (Leenders et al. 1996b, Adamski et al. 1997). Also are predominantly seen in the placenta. the preferred reaction direction, i.e. oxidation, Several human cancer cell lines also express remained unchanged for purified, recombinant and 17â-HSD 4. The estrogen receptor positive mam- transiently expressed proteins. mary cell line, T47D, expresses more 17â-HSD 4 mRNA transcript than BT-20, MDA-MB-453 and MDA-MB-231 cell lines which are estrogen TISSUE DISTRIBUTION receptor negative. The megakaryotic cell line, DAMI, reveals a very high level of 17â-HSD 4 In the porcine tissues studied, the oxidation of mRNA while less is present in hepatocellular 17â-estradiol predominates over the reduction. carcinoma HEP-G2 and early embryonic Tera-1

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   233 cell lines (Adamski et al. 1995). The prostate cancer a down-regulation (instead of up-regulation) of the cell lines DU145, LNCaP and PC3 express expression of 17â-HSD 4 (Caira et al. 1998). 17â-HSD 4 but not 17â-HSD 1 or 3 (Carruba et al. 1997, Castagnetta et al. 1997). Interestingly, 17â-HSD 2, which appears to be the principal SUBCELLULAR DISTRIBUTION OF PORCINE isozyme expressed in the prostate, is present only in 17â-HSD 4 PC3 cells (Delos et al. 1995, 1998, Elo et al. 1996, Castagnetta et al. 1997). Most probably, 17â-HSD Immunocytochemical and immunofluorescence 2 is the main protective dehydrogenase in the studies in porcine uterus restricted porcine 17â- prostate. HSD 4 to luminal and glandular epithelium (Husen et al. 1994) similar to analyses of human endo- metrium (Scublinsky et al. 1976, Ma¨entausta et al. REGULATION OF HSD17B4 1991). However, the staining in the cytoplasm was not diffuse but showed a punctuate appearance. The Porcine 17â-hydroxysteroid dehydrogenase 4 is the intensity of the monoclonal antibody F1-peroxidase first peroxisomal enzyme known to be stimulated by staining followed the changes in porcine 17â- progesterone as checked by mRNA expression and hydroxysteroid dehydrogenase activity. It was immunohistochemistry (Kaufmann et al. 1995). In raised fourfold after day 5 of the ovarian cycle and the early 1970s the hormone was observed to rapidly decreased after day 17 in a manner similar to increase estradiol oxidation in human tissues (Tseng the levels of progesterone. On day 4 faint spots of & Gurpide 1975, 1979). Later it was shown to fluorescence appeared in the cytoplasm of the up-regulate 17â-HSD 1 in human breast cancer glandular epithelium. The spots accumulated at cells (Poutanen et al. 1990) and to increase mRNA the cell bases between days 11 and 17 (luteal phase) expression of 17â-HSD 2 in human endometrium and disappeared within one day. The pattern of (Casey et al. 1994). immunofluorescence staining suggested that porcine Although 17â-HSD 1 is supposed to participate 17â-HSD 4 is localized in vesicles. The latter have in the synthesis and 17â-HSD 4 in the inactivation been isolated from porcine uterus epithelium of steroids there is a simultaneous expression of homogenates by sequential density gradients of both enzymes in gonads (Luu-The et al. 1990, isopycnic 30% Percoll and linear 0·3–2 M sucrose in Carstensen et al. 1996). However, the correspond- vertical rotors (Adamski et al. 1987, Adamski 1991, ing regulatory pathways of protein kinase C are Adamski et al. 1993). The vesicles harboring the not controlled in the same way. In vitamin 17â-HSD activity equilibrated at a density of D-differentiated human leukemia THP 1 cells, 1·18 g/ml, were 120–200 nm in diameter, revealed 17â-HSD 4 mRNA was stimulated twofold by a moderate electron–dense matrix bounded by a dexamethasone but it was completely down- single membrane and were morphologically and regulated by phorbol esters (Jakob et al. 1995, enzymatically distinct from mitochondria, lyso- 1997). This is in contrast to the dose- and somes, fragments of plasma membrane, endoplas- time-dependent increase in gene transcripts of mic reticulum and the Golgi apparatus. In 17â-HSD 1 under similar treatment (Tremblay & immunogold electron microscopy the labeling with Beaudoin 1993). monoclonal antibody F1 (recognizing the 32 kDa The recently purified and cloned rat 80 kDa and the 80 kDa protein) and W1 (reacting with homolog of human 17â-HSD 4 is up-regulated by 32 kDa only) confirmed that all forms of the enzyme peroxisomal proliferators such as clofibrate and WY are present in the same vesicles, both in tissue and 14,643 (Novikov et al. 1994, Corton et al. 1995). In in the isolated fraction (Adamski et al. 1993). the PPARá (peroxisomal-proliferator activated re- Several clues pointed to the identity of the ceptor alpha) knock-out mice the expression of 17â-HSD 4 containing vesicles as peroxisomes: (1) 17â-HSD 4 is low and does not change after the morphology and density is similar to that of treatment with WY 14,643 (Aoyama et al. 1998). peroxisomes, (2) the 80 kDa primary transcript The 80 kDa protein seems to be controlled by features the peroxisomal targeting signal Ala-Lys- modulators of both steroid and fatty acid metab- Ile and a putative recognition sequence (Ala-Ala- olism. Another example of PPARá-mediated regu- Pro) for a protease processing peroxisomal protein lation is the activation of steroid metabolizing (Mori et al. 1991) and (3) the 80 kDa protein is enzyme, NADP+-dependent 3á-HSD in human similar to enzymes participating in peroxisomal liver by derivatives of clofibrate (Matsuura et al. â-oxidation of fatty acids. Indeed, typical peroxi- 1996). In contrast, the guinea pig ortholog is so far somal markers such as and acyl-CoA the only species in which the same treatment causes oxidase co-localized with porcine 17â-HSD 4 in

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 234    and   · 17â-HSD 4 ) immunogold labeling studies in uterus, kidney and 1 liver (Markus et al. 1995a,b). " In other species, such as the rat, 17â-HSD 4 was indeed purified from isolated peroxisomes (Novikov . 1996). mg prot. et al

et al. 1994). However, at that time the amino acid # 1

sequence of the rat enzyme was not known. " Peroxisomal localization of 17â-HSD 4 extends min

our understanding on how steroid, sterol and bile # 1

acid are interlinked. Different sub- " l

cellular distribution of various 17â-HSDs allows # for local control of their activities. Peroxisomes mol max ì . 1996). The recombinant central domain ( were initially believed to play only a minor role in V

mammalian metabolism. However, they play an et al M) indispensable role in many metabolic pathways, like m ì K ( synthesis of cholesterol, â-oxidation of fatty acids, Fatty acid-CoA dehydrogenase

biosynthesis of ether lipids and bile acids, ) 1

á-oxidation of phytanic acid and, as depicted in this " review, oxidation of steroids (Wanders et al. 1995, Krisans 1996, Magalhaes & Magalhaes 1997,

Verhoeven et al. 1997, Seedorf et al. 1998). mg prot. # 1 "

MULTIFUNCTIONALITY OF 17â-HSD 4 min # 1 " In order to check for the presence of activities l predicted by amino acid similarities of the 80 kDa # mol protein (Fig. 2) its three domains were expressed max ì ( separately (Leenders et al. 1996b). The N-terminal V domain (aa 1–323) catalyzed both the 17â- M) m ì ( K hydroxysteroid and the 3-hydroxyacyl-CoA dehy- Fatty acid-CoA hydratase drogenase reactions (Table 5). Kinetic parameters ) (Km,Vmax) of the expressed full length 80 kDa 1 protein were close to those observed for the single " expressed domains or for the native purified enzyme (EDH or VHF) (Leenders et al. 1996b). mg prot. # This was the first observation of an enzyme -HSD 4 1 â

performing dehydrogenase activity not only with " steroids but also with 3-hydroxyacyl-CoA derivates min

of fatty acids. Furthermore, the central domain # 1 -estradiol, fatty acid-CoA hydratase with crotonyl-CoA and fatty acid-CoA dehydrogenase with acetoacetyl-CoA (Leenders "

(aa 324–596) was responsible for the 2-enoyl-acyl- l â

CoA hydratase activity. Essentially the same data # concerning conversion of 3-hydroxyacyl-CoAs and max (nmol 2-enoyl-acyl-CoAs were obtained simultaneously V with expressed or purified proteins of the rat -Hydroxysteroid â M) (Novikov et al. 1994, Qin et al. 1997a,b). Estimated m ì K dehydrogenase 17 ( Km values for the hydratase and fatty acid dehydrogenase are similar to those of other enzymes of â-oxidation of fatty acids (Steinman & Hill 1975, -HSD 4 were assayed with 17

Palosaari & Hiltunen 1990). Both Km for steroids â and fatty acids are in the physiological range. Further studies on peroxisomal â-oxidation of fatty acids and precursors of bile acids demon- strated that the 17â-HSD 4 specifically forms and dehydrogenates -3-hydroxyacyl-CoAs and not 5. Kinetic parameters of domains of porcine 17 their -stereoisomers (Dieuaide-Noubhani et al. Sample Purified 32 kDaRecombinant 32 kDaVHFRecombinant 80 kDaRecombinant central domain 0·3 ND 0·2 ND 0·4 0·14 0·15 0·11 0·3 0·19 34·7 ND ND 1·36 ND 37·1 ND 3·92 34·0 4·44 21·9 ND ND 1·25 ND 35·3 ND 2·91 34·8 3·31  was characterized after purificationND, on activity glutathione-agarose not and detected. thrombin cleavage. Recombinant 32 kDa and 80 kDa proteins were assayed in homogenates of transfected cells and were corrected for background conversion (Carstensen 1997a, Jiang et al. 1997). This differentiates the Kinetic parameters of 17

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   235 enzyme from the multifuntional enzyme 1 (MFP1) The role of SCP2 in steroidogenesis and arterio- (Osumi et al. 1985b) which is -specific. sclerosis has been extensively studied (Seedorf et al. The velocity of 17â-estradiol oxidation by 1993, Krisans 1996, Magalhaes & Magalhaes porcine, mouse and human 17â-HSD 4 is several 1997, Wanders et al. 1997). A synopsis of this fold lower than that of fatty acyl-CoA (Leenders functionality is beyond the scope and limits of this et al. 1994a, 1996b, Adamski et al. 1995, 1997, review. Normand et al. 1995). The same observation was The role of the SCP2 domain for the functionality made in rat (Dieuaide-Noubhani et al. 1996b, Qin of 17â-HSD 4 is not clear. The expressed por- et al. 1997b). However, all other known 17â-HSDs cine SCP2 domain facilitates the transfer of have the same conversion rates for steroids within 7-dehydrocholesterol and phosphatidylcholine be- the range 0·1 to 2·45 nmol/min/mg protein (Table tween membranes in vitro. The activities of the 1). Because the enzymatic parameters (Vmax, Km) N-terminal domain towards steroids or fatty of the 17â-HSD 4 for both fatty acyl-CoA and acyl-CoA are not changed if the SCP2 domain is steroids are close to those known for other enzymes deleted (Leenders et al. 1996b). of the SCAD gene family it remains to be settled Recently, gene targeting in mice was used to which substrates are physiological. study the unknown function of SCP2 (Seedorf et al. In comparison to other 17â-HSDs the type 4 1998). In the Scp2(-/-) mice with complete enzyme accepts the most structurally diverse deficiency of SCP2 and SCPX, marked alterations substrates. Human 17â-HSD 1–3 and mouse and in , proliferation, hypo- rat 17â-HSD 7 (Nokelainen et al. 1998) are lipidemia, impaired body weight control, and practically only active with steroids at position 17. neuropathy were observed. Knock-out mice showed However, 17â-HSD 5 and the recently cloned rat impaired catabolism of methyl-branched fatty 17â-HSD 6 (Biswas & Russell 1997) must be acyl-CoAs, especially of the tetramethyl-branched considered multifunctional because of their 3á- fatty acid, phytanic acid. The gene disruption led to hydroxysteroid dehydrogenase activity. Actually, inefficient import of phytanoyl-CoA into peroxi- 17â-HSD 5 was first identified in a row of different somes and to defective thiolytic cleavage of 3á-HSDs and assigned type 2 among them (for 3-ketopristanoyl-CoA. reviews see Lin et al. 1997, Penning 1997). One 3á-HSD was even first identified as a bile acid binding protein (Nanjo et al. 1995). MUTATIONS IN THE HSD17B4 GENE Interesting is the novel 17â-hydroxysteroid dehydrogenase previously known as Ke6 protein in Recent research on peroxisomal disorders revealed mouse and human (Ando et al. 1996, Formicheva that 17â-HSD 4 is deficient in Zellweger syndrome et al. 1998). This enzyme which might be termed (Novikov et al. 1997, Suzuki et al. 1997, van 17â-HSD 8 has a Vmax of 0·27 nmol/min/mg Grunsven et al. 1998). Peroxisomal organelle protein with 17â-estradiol/NAD+ and reveals the deficiency results in disorders of lipid, fatty acid and highest (37%) amino acid sequence identity among sterol metabolism such as Zellweger syndrome, the 17â-HSDs to 17â-HSD 4. It remains to be adrenoleukodystrophy, infantile refsum disease and verified if 17â-HSD 8 is able to metabolize bile hyperpipecolic acidemia (Lazarow & Moser 1995, acids or fatty acids. Wanders et al. 1995). Patients with peroxisomal deficiency reveal high plasma concentrations of long chain fatty acids, bile acids and deficient synthesis ROLE OF SCP2 DOMAIN of plasmalogens. This exerts pleiotropic influence of renal functions impairment and neuronal The most C-terminal part of the 80 kDa protein development (neuronal migration defects and (amino acids 597–737) has 39% similarity to rat degeneration). SCP2 (Leenders et al. 1994b). This non-specific One example of the molecular basis of the lipid transfer protein is highly conserved, even in recently identified 17â-HSD 4 deficiency is the evolutionarily distant species such as chicken and mutation G16S (van Grunsven et al. 1998). This human (Yamamoto et al. 1991, Pfeifer et al. 1993). mutation is localized in the first exon (Fig. 2) and This product of an SCPX gene, which actually disturbs the conformation of the Rossman fold encodes two proteins, SCP2 and SCPX, is a fusion required for cofactor (NAD+) binding. The mutant between SCP2 and thiolase (Ohba et al. 1994, is inactive with both steroids and bile acids (van Seedorf et al. 1994). SCP2 is a 13 kDa basic Grunsven et al. 1998, Mo¨ller et al. 1999). The protein believed to participate in the intracellular mutation is lethal, most probably because movement of cholesterol and lipids (Wirtz 1997). the -specific pathways of pristanic acid and

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 236    and   · 17â-HSD 4

di/tri-hydroxycholestanoic acid metabolism are (Grandien et al. 1995). 17â-HSD 4 inactivates the disrupted. As mentioned above, the multifunctional conversion of Ä5-androstene-3â,17â-diol to dehy- protein 1 (Osumi et al. 1985a)is-specific and droepiandrosterone (DHEA), a known peroxisomal cannot substitute the deficient -pathway proliferator (Prough et al. 1994). Although both (Dieuaide-Noubhani et al. 1997a). Because 17â- DHEA and clofibrate induce peroxisomes they HSD 4 has a ubiquitous distribution, any mutation have opposite effects on the concentrations of would affect the whole organism. In addition, triglycerides and cholesterol in blood. DHEA developmental studies have shown that this enzyme increases the levels of lipids while clofibrate acts as is present as early as at least day 7 post coitus of a hyperlipidemic drug. Decreased expression of embryonic development (Mustonen et al. 1997). enzymes which inactivate estradiol, including The lack of observation of any steroid hormone Cyp2C11, and the reported increased expression of related phenotype might be due to compensation by aromatase (converting testosterone to estradiol) may other 17â-hydroxysteroid dehydrogenases. explain why male rats exposed to diverse peroxi- somal proliferators have higher serum estradiol levels. These higher estradiol levels in male rats PHYSIOLOGICAL SIGNIFICANCE IN have been thought to be mechanistically linked to STEROID METABOLISM Leydig cell hyperplasia and adenomas. Increased conversion of estradiol to the less active estrone by Table 1 compares human 17â-HSD 1–5 and depicts 17â-HSD 4 induction may explain how exposure to differences in catalytic parameters, posttranslational the di-(2-ethylhexyl)-phthalate leads to decreases in processing and subcellular localization. The cata- serum estradiol levels and suppression of ovulation lytical property of 17â-HSD 4, revealing the in female rats (Srivastava & Srivastava 1991, Corton virtually unidirectional oxidative activity, clearly et al. 1997). defines it as a steroid inactivating enzyme (Gurpide & Marks 1981), since it produces estrone which shows little affinity to the estradiol receptor. The ACKNOWLEDGEMENTS conversion of 17â-estradiol to estrone might be complemented by hydroxylations in positions 6á or The authors did their best to review known data on 7á (Maschler et al. 1983) producing steroids devoid 17â-HSD 4 within this review. We apologize to our ffi of estradiol receptor a nity and permitting fast colleagues whose contributions were not included release from cells after formation. The Vmax and Km here. This occurred solely because of limitation of values for EDH are similar to those for estrone space. hydroxylases (Adamski et al. 1994) and allows the This work has been carried out on the basis of metabolic conversion 17â-estradiol

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   237

localized in specialized vesicles. Biochemical Journal 290 Corton JC, Bocos C, Cattley RC & Gustafsson JA 1995 The 777–782. rat 17â-estradiol dehydrogenase type IV is a novel Adamski J, HohlsE&Jungblut PW 1994 Characterization of peroxisome proliferator responsive gene. In Peroxisomes: estrone hydroxylase activities in procine endometrial cells. Biology and Role in Toxicology and Disease. Aspen, Colorado, Experimental and Clinical Endocrinology 102 388–393. USA: The Aspen Institute. Adamski J, Normand T, Leenders F, Monte D, Begue A, Corton JC, Bocos C, Moreno ES, Merritt A, Marsman DS, Stehelin D, Jungblut PW & de Launoit Y 1995 Molecular Sausen PJ, Cattley RC & Gustafsson JA 1996 The rat cloning of a novel widely expressed human 80 kDa 17â-hydroxysteroid dehydrogenase type IV is a novel 17â-hydroxysteroid dehydrogenase IV. Biochemical Journal peroxisome-proliferator-inducible gene. Molecular 311 437–443. Pharmacology 50 1157–1166. Adamski J, Leenders F, Carstensen JF, Kaufmann M, Markus Corton JC, Bocos C, Moreno ES, Merritt A, Cattley RC & MM, Husen B, Tesdorpf JG, Seedorf U, de Launoit Y & Gustafsson JA 1997 Peroxisome proliferators alter the Jakob F 1997 Steroids, fatty acyl-CoA, and sterols are expression of estrogen-metabolizing enzymes. Biochimie 79 substrates of 80-kDa multifunctional protein. Steroids 62 151–162. 159–163. Delos S, Carsol JL, Ghazarossian E, Raynaud JP & Martin PM Andersson S 1995 17â-Hydroxysteroid dehydrogenases: 1995 Testosterone metabolism in primary cultures of human isozymes and mutations. Journal of Endocrinology 146 prostate epithelial cells and fibroblasts. Journal of Steroid 197–200. Biochemistry and Molecular Biology 55 375–383. Ando A, Kikuti YY, Shigenari A, Kawata H, Okamoto N, Delos S, Carsol JL, Fina F, Raynaud JP & Martin PM 1998 Shiina T, Chen L, Ikemura T, Abe K, Kimura M & Inoko 5á-Reductase and 17â-hydroxysteroid dehydrogenase H 1996 cDNA cloning of the human homologues of the expression in epithelial cells from hyperplastic and mouse Ke4 and Ke6 genes at the centromeric end of the malignant human prostate. International Journal of Cancer 75 human MHC region. Genomics 35 600–602. 840–846. Aoyama T, Peters JM, Iritani N, Nakajima T, Furihata K, Deyashiki Y, Ohshima K, Nakanishi M, Sato K, Matsuura K Hashimoto T & Gonzalez FJ 1998 Altered constitutive & Hara A 1995 Molecular cloning and characterization of expression of fatty acid-metabolizing enzymes in mice mouse estradiol 17 beta-dehydrogenase (A-specific), a lacking the peroxisome proliferator-activated receptor alpha member of the aldoketoreductase family. Journal of Biological (PPARá). Journal of Biological Chemistry 273 5678–5684. Chemistry 270 10461–10467. Baker ME 1994 Sequence analysis of steroid- and Dieuaide-Noubhani M, Novikov D, Baumgart E, Vanhooren prostaglandin-metabolizing enzymes: application to JC, Fransen M, Goethals M, Vandekerckhove J, understanding catalysis. Steroids 59 248–258. Van Veldhoven PP & Mannaerts GP 1996a Further Baker ME 1996 Unusual evolution of 11beta- and 17beta- characterization of the peroxisomal 3-hydroxyacyl-CoA hydroxysteroid and retinol dehydrogenases. Bioessays 18 dehydrogenases from rat liver. Relationship between the 63–70. different dehydrogenases and evidence that fatty acids and Biswas MG & Russell DW 1997 Expression cloning and the C27 bile acids di- and tri-hydroxycoprostanic acids are characterization of oxidative 17beta- and 3alpha- metabolized by separate multifunctional proteins. European hydroxysteroid dehydrogenases from rat and human prostate. Journal of Biochemistry 240 660–666. Journal of Biological Chemistry 272 15959–15966. Dieuaide-Noubhani M, Novikov D, Baumgart E, Vanhooren Blomquist CH, Lindemann NJ & Hakanson EY 1985 JCT, Fransen M, Goethals M, Vandekerkhove J, 17â-Hydroxysteroid and 20á-hydroxysteroid dehydrogenase Van Veldhoven PP & Mannaerts GP 1996b Further activities of human placental microsomes: kinetic evidence characterisation of the peroxisomal 3-hydroxyacyl-CoA for two enzymes differing in substrate specificity. Archives of dehydrogenase from rat liver. Relationship between different Biochemistry and Biophysics 239 206–215. dehydrogenases and evidence that fatty acids and the C27 Caira F, Clemencet MC, Cherkaoui-Malki M, Dieuaide- bile acids di- and tri-hydroxycoprostanic acids are Noubhani M, Pacot C, Van Veldhoven PP & LatruffeN metabolized by separate multifunctional proteins. European 1998 Differential regulation by a peroxisome proliferator of Journal of Biochemistry 240 660–666. the different multifunctional proteins in guinea pig: cDNA Dieuaide-Noubhani M, Asselberghs S, Mannaerts GP & Van cloning of the guinea pig -specific multifunctional protein 2. Veldhoven PP 1997a Evidence that multifunctional protein Biochemical Journal 330 1361–1368. 2, and not multifunctional protein 1, is involved in the Carruba G, Adamski J, Calabro` M, Miceli MD, Cataliotti A, peroxisomal beta-oxidation of pristanic acid. Biochemical Bellavia V, Bue AL, Polito L & Castagnetta L 1997 Molecular Journal 325 367–373. expression of 17â-hydroxysteroid dehydrogenase types in Dieuaide-Noubhani M, Novikov D, Vandekerckhove J, relation to their activity in intact human prostate cancer cells. Veldhoven PPV & Mannaerts GP 1997b Identification and Molecular and Cellular Endocrinology 131 51–57. characterization of the 2-enoyl-CoA hydratases involved in Carstensen JF, Tesdorpf JG, Kaufmann M, Markus MM, peroxisomal beta-oxidation in rat liver. Biochemical Journal Husen B, Leenders F, Jakob F, de Launoit Y & Adamski J 321 253–259. 1996 Characterization of 17â-hydroxysteroid dehydrogenase Elo JP, Akinola LA, Poutanen M, Vihko P, Kyllonen AP, IV. Journal of Endocrinology 150 3–12. LukkarinenO&VihkoR1996Characterization of Casey ML, MacDonald PC & Andersson S 1994 17â- 17â-hydroxysteroid dehydrogenase isoenzyme expression in Hydroxysteroid dehydrogenase type 2: chromosomal benign and malignant human prostate. International Journal assignment and progestin regulation of gene expression in of Cancer 66 37–41. human endometrium. Journal of Clinical Investigation 94 Engel LL & Groman EV 1974 Human placental 17â-estradiol 2135–2141. dehydrogenase: characterization and structual studies. Recent Castagnetta L, Carruba G, Traina A, Granata OM, Markus M, Progress in Hormone Research 30 139–169. Pavone-Macaluso M, Blomquist CH & Adamski J 1997 Entenmann AH, Sierralta WD & Jungblut PW 1980 The Expression of different 17â-hydroxysteroid dehydrogenase dehydrogenation of estradiol to estrone by porcine types and their activity in human prostate cancer cells. endometrial lysosomes. Hoppe-Seyler’s Zeitschrift fu¨r Endocrinology 138 4876–4882. Physiologische Chemie 361 959–968.

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 238    and   · 17â-HSD 4

Formicheva J, Baker M, Anderson E, Lee GY & Aziz N 1998 Lazarow PB & Moser HW 1995 Disorders of Peroxisome Characterization of Ke6, a new 17â-hydroxysteroid Biogenesis. New York: McGraw-Hill. dehydrogenase and its expression in gonadal tissues. Journal Leenders F, Adamski J, Husen B, Thole HH & Jungblut PW of Biological Chemistry 273 22664–22671. 1994a Molecular cloning and amino acid sequence of the Geissler WM, Davis DL, Wu L, Bradshaw KD, Patel S, porcine 17â-estradiol dehydrogenase. European Journal of Mendonca BB, Elliston KO, Wilson JD, Russell DW & Biochemistry 222 221–227. Andersson S 1994 Male pseudohermaphroditism caused by Leenders F, Husen B, Thole HH & Adamski J 1994b mutations of testicular 17â-hydroxysteroid dehydrogenase 3. The sequence of porcine 80 kDa 17â-estradiol Nature Genetics 7 34–39. dehydrogenase reveals similarities to the short chain alcohol Grandien K, Backdahl M, Ljunggren O, Gustafsson JA & dehydrogenase family, to actin binding motifs and to sterol Berkenstam A 1995 Estrogen target tissue determines carrier protein 2. Molecular and Cellular Endocrinology 104 alternative promoter utilization of the human estrogen 127–131. receptor gene in osteoblasts and tumor cell lines. Leenders F, Prescher G, de Launoit Y & Adamski J 1996a Endocrinology 136 2223–2229. Assignment of human 17â-hydroxysteroid dehydrogenase IV van Grunsven EG, van Berkel E, Ijlst L, Vreken P, de Klerk to chromosome 5q2 by fluorescence in situ hybridization. JBC, Adamski J, Lemonde H, Clayton PT, Cuebas DA & Genomics 37 403–404. Wanders RJA 1998 Peroxisomal -hydroxyacyl-CoA Leenders F, Tesdorpf JG, Markus M, Engel T, Seedorf U & dehydrogenase deficiency: resolution of the enzyme defect Adamski J 1996b Porcine 80 kDa protein reveals intrinsic and its molecular basis in bifunctional protein deficiency. 17â-hydroxysteroid dehydrogenase, fatty acyl-CoA- Proceedings of the National Academy of Sciences of the USA hydratase/dehydrogenase and sterol transfer activities. 95 2128–2133. Journal of Biological Chemistry 271 5438–5442. GurpideE&MarksC1981 Influence of endometrial 17â- Leenders F, Dolez V, Begue A, Mo¨ller G, Gloeckner JC, de hydroxysteroid dehydrogenase activity on the binding of Launoit Y & Adamski J 1999 Structure of the gene for the estradiol to receptors. Journal of Clinical Endocrinology and human 17â-hydroxysteroid dehydrogenase type IV. Metabolism 52 252–255. Mammalian Genome 9 1036–1041. Husen B, Adamski J, Szendro PI & Jungblut PW 1994 Lin HK, Jez JM, Schlegel BP, Peehl DM, Pachter JA & Alterations in the subcellular distribution of 17â-estradiol Penning TM 1997 Expression and characterization of dehydrogenase in porcine endometrial cells over the recombinant type 2 3á-hydroxysteroid dehydrogenase (HSD) course of the estrous cycle. Cell and Tissue Research 278 from human prostate: demonstration of bifunctional 227–233. 3á/17â-HSD activity and cellular distribution. Molecular Jakob F, Homann D & Adamski J 1995 Expression and Endocrinology 11 1971–1984. regulation of aromatase and 17â-hydroxysteroid Luu-The V, Lachance Y, Labrie C, Leblanc G, Thomas JL, dehydrogenase type 4 in human THP 1 leukemia cells. Strickler RC & Labrie F 1989 Full length cDNA structure Journal of Steroid Biochemistry and Molecular Biology 55 and deduced amino acid sequence of human 3â-hydroxy- 555–563. 5-ene steroid dehydrogenase. Molecular Endocrinology Jakob F, Siggelkow H, Homann D, Kohrle J, Adamski J & 3 1310–1312. Schutze N 1997 Local estradiol metabolism in osteoblast- Luu-The V, Labrie C, Simard J, Lachance Y, Zhao H-F, and osteoclast-like cells. Journal of Steroid Biochemistry and CouetJ,LeblancG&LabrieF1990 Structure of two in Molecular Biology 61 167–174. tandem human 17â-hydroxysteroid dehydrogenase genes. Jiang LL, Kobayashi A, Matsuura H, Fukishima H & Molecular Endocrinology 4 268–275. Hashimoto T 1996 Purification and properties of human Ma¨entausta O, Sormunen R, Isomaa V, Lehto V-P, Jouppila P -3-hydroxyacyl-CoA dehydratase: medium chain enoyl-CoA & Vihko R 1991 Immunohistochemical localization of hydratase is -3-hydroxyacyl-CoA dehydratase. Journal of 17â-hydroxysteroid dehydrogenase in the human Biochemistry 120 624–632. endometrium during the menstrual cycle. Laboratory Jiang LL, Kurosawa T, Sato M, SuzukiY&HashimotoT Investigation 65 582–587. 1997 Physiological role of -3-hydroxyacyl-CoA dehydratase/ Magalhaes MM & Magalhaes MC 1997 Peroxisomes in adrenal -3-hydroxyacyl-CoA dehydrogenase bifunctional protein. steroidogenesis. Microscopy Research and Technique 36 Journal of Biochemistry 121 506–513. 493–502. Jo¨rnvall H, Persson B, Krook M, Atrian S, Gozales-Duarte R, Marks F 1992 Isolierung und Charakterisierung der 17â- Jeffery J & Ghosh D 1995 Short-chain dehydrogenases/ Estradioldehydrogenase des Schweineuterus. PhD Thesis, reductases (SDR). Biochemistry 34 6003–6013. University of Hannover, Germany. Kaufmann M, Carstensen J, Husen B & Adamski J 1995 Markus M, Husen B & Adamski J 1995a The subcellular The tissue distribution of porcine 17â-estradiol localization of 17â-hydroxysteroid dehydrogenase type 4 and dehydrogenase and its induction by progesterone. its interaction with actin. Journal of Steroid Biochemistry and Journal of Steroid Biochemistry and Molecular Biology 55 Molecular Biology 55 617–621. 535–539. Markus M, Husen B, Leenders F, Jungblut PW, Hall PF & Kobayashi K, Kobayashi H, Ueda M & Honda Y 1997 Adamski J 1995b The organelles containing porcine 17â- Expression of 17â-hydroxysteroid dehydrogenase type IV in hydroxysteroid dehydrogenase are peroxisomes. European chick retinal pigment epithelium. Experimental Eye Research Journal of Cell Biology 68 263–267. 64 719–726. Martel C, Rheaume E, Takahashi M, Trudel C, Couet J, Luu Krisans SK 1996 Cell compartmentalization of cholesterol TV,SimardJ&LabrieF1992Distribution of 17 beta- biosynthesis. Annals of the New York Academy of Sciences hydroxysteroid dehydrogenase gene expression and activity 804 142–164. in rat and human tissues. Journal of Steroid Biochemistry and Labrie Y, Durocher F, Lachance Y, Turgeon C, Simard J, Molecular Biology 41 597–603. LabrieC&LabrieF1995 The human type II 17â- Maschler I, Ball P, Bayerko¨hler G, Gaues J & Knuppen R hydroxysteroid dehydrogenase gene encodes two 1983 Identification of 6á-and7á-hydroxyestrone as major alternatively spliced mRNA species. DNA and Cell Biology metabolites of estrone and estradiol in porcine uterus. 14 849–861. Steroids 41 597–607.

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 17â-HSD 4 ·    and   239

Matsuura K, Tamada Y, Deyashiki Y, Miyabe Y, Nakanishi 2-enoyl-CoA hydratase, 3-hydroxy-CoA dehydrogenase and M,OhyaI&HaraA1996 Activation of human liver 3,2-enoyl-CoA isomerase activities. Journal of Biological 3á-hydroxysteroid dehydrogenase by sulphobromophthalein. Biochemistry 265 2446–2449. Biochemical Journal 313 179–184. Peltoketo H, Isomaa V, Ma¨entaustaO&VihkoR1988 Mo¨ller G, Leenders F, van Grunsven E, Dolez V, Qualmann Complete amino acid sequence of human placental B, Kessels MM, Markus M, Krazeisen A, Husen B, 17â-hydroxysteroid dehydrogenase deduced from cDNA. Wanders RJA, de Launoit Y & Adamski J 1999 FEBS Letters 239 73–77. Characterization of the HSD17B4 gene: -specific Penning TM 1997 Molecular endocrinology of hydroxysteroid multifunctional protein 2/17â-hydroxysteroid dehydrogenase dehydrogenases. Endocrine Reviews 18 281–305. IV. Journal of Steroid Biochemistry and Molecular Biology (In Penning TM, Bennett MJ, Smith-Hoog S, Schlegel BP, Jez Press). JM & Lewis M 1997 Structure and function of 3á- Mori T, Tsukamoto T, Mori H, Tashiro Y & Fujiki Y 1991 hydroxysteroid dehydrogenase. Steroids 62 101–111. Molecular cloning and deduced amino acid sequence of Persson B, Krook M & Jo¨rnvall H 1991 Characteristics of nonspecific lipid transfer protein (sterol carrier protein 2) of short-chain alcohol dehydrogenases and related enzymes. rat liver: a higher molecular mass (60 kDa) protein contains European Journal of Biochemistry 200 537–543. the primary sequence of nonspecific lipid transfer protein as Pfeifer SM, Sakuragi N, Ryan A, Johnson AL, Deeley RG, its C-terminal part. Proceedings of the National Academy of Billheimer JT, Baker ME & Strauss JF 1993 Chicken sterol Sciences of the USA 88 4338–4342. carrier protein 2/sterol carrier protein x: cDNA cloning Mustonen MVJ, Poutanen MH, Isomaa VV, Vihko PT & reveals evolutionary conservation of structure and regulated Vihko RK 1997 Cloning of mouse 17â-hydroxysteroid expression. Archives of Biochemistry and Biophysics 304 dehydrogenase type 2, and analysing expression of the 287–293. mRNAs for types 1, 2, 3, 4 and 5 in mouse embryos and Pollow K, Lu¨bbert H & Pollow B 1976 Partial purification and adult tissues. Biochemical Journal 325 199–205. evidence of heterogeneity of the cytoplasmic 17â- Nanjo H, Adachi H, Morihana S, Mizoguchi T, Nishihara T & hydroxysteroid dehydrogenase (17â-HSD) from normal Terada T 1995 Enzymatic characterization of a novel bovine human endometrium and endometrial carcinoma. Journal of liver dihydrodiol dehydrogenase – reaction mechanism and Steroid Biochemistry 7 315–320. bile acid dehydrogenase activity. Biochimica et Biophysica Poutanen M, Isomaa V, KainulainenK&VihkoR1990 Acta 1244 53–61. Progestin induction of 17â-hydroxysteroid dehydrogenase Nokelainen P, Peltoketo H, VihkoR&VihkoP1998 enzyme protein in the T-47D human breast-cancer cell line. Expression cloning of a novel estrogenic mouse 17â- International Journal of Cancer 46 897–901. hydroxysteroid dehydrogenase/17-ketosteroid reductase Poutanen M, Isomaa V, PeltoketoH&VihkoR1995 (m17HSD7), previously described as a prolactin receptor- Regulation of oestrogen action: role of 17â-hydroxysteroid associated protein (PRAP) in rat. Molecular Endocrinology 12 dehydrogenases. Annals of Medicine 27 675–682. 1048–1059. Prough RA, Webb SJ, Wu HQ, Lapenson DP & Waxman DJ Normand T, Husen B, Leenders F, Pelczar H, Baert JL, Begue 1994 Induction of microsomal and peroxisomal enzymes by A,FlourensAC,AdamskiJ&deLaunoit Y 1995 Molecular dehydroepiandrosterone and its reduced metabolite in rats. characterization of mouse 17â-hydroxysteroid dehydrogenase Cancer Research 54 2878–2886. IV. Journal of Steroid Biochemistry and Molecular Biology 55 Qin YM, Haapalainen AM, Conry D, Cuebas DA, Hiltunen 541–548. JK & Novikov DK 1997a Recombinant 2-enoyl-CoA Novikov DK, Vanhove G, Carchon H, Asselberghs S, Eyssen hydratase derived from rat peroxisomal multifunctional HJ, Van Veldhoven PP & Mannaerts GP 1994 Peroxisomal enzyme 2: role of the hydratase reaction in bile acid â-oxidation. Purification of four novel 3-hydroxyacyl-CoA synthesis. Biochemical Journal 328 377–382. dehydrogenases from rat liver peroxisomes. Journal of Qin YM, Poutanen MH, Helander HM, Kvist AP, Siivari Biological Chemistry 269 27125–27135. KM, Schmitz W, Conzelmann E, HellmanU&Hiltunen JK Novikov D, Dieuaide-Noubhani M, Vermeesch JR, Fournier 1997b Peroxisomal multifunctional enzyme of beta-oxidation B, Mannaerts GP & Van Veldhoven PP 1997 The human metabolizing -3-hydroxyacyl-CoA esters in rat liver: peroxisomal multifunctional protein involved in bile acid molecular cloning, expression and characterization. synthesis: activity measurement, deficiency in Zellweger Biochemical Journal 321 21–28. syndrome and chromosome mapping. Biochimica et Reed MJ 1991 Oestradiol-17â hydroxysteroid dehydrogenase: Biophysica Acta 1360 229–240. its family and function. Journal of Endocrinology 129 Ohba T, Rennert H, Pfeifer SM, He Z, Yamamoto R, Holt JA, 163–165. Bilheimer JT & Strauss III JF 1994 The structure of the Scublinsky A, Marin C & Gurpide E 1976 Localization of human sterol carrier protein x/ sterol carrier protein 2 estradiol 17â dehydrogenase in human endometrium. Journal (SCP2) gene. Genomics 24 370–374. of Steroid Biochemistry 7 745–747. Osumi T, Ishii N, Hijikata M, Kamijo K, Ozasa H, Furuta S, Seedorf U, Raabe M & Assmann G 1993 Cloning, expression Miyazawa S, Kondo K, Inoue K, Kagamiyama H et al. and sequences of mouse sterol carrier protein-X encoding 1985a Molecular cloning and nucleotide sequence of the cDNAs and related pseudogene. Gene 123 165–172. cDNA for rat peroxisomal enoyl-CoA: hydratase-3- Seedorf U, Brysch P, Engel T, SchrageK&AssmannG hydroxyacyl-CoA dehydrogenase bifunctional enzyme. 1994 Sterol carrier protein-X is a peroxisomal 3-oxoacyl Journal of Biological Chemistry 260 8905–8910. coenzyme A thiolase with intrinsic sterol carrier and lipid Osumi T, Ishii N, Hijikata M, Kamijo K, Ozasa H, Furuta S, transfer activity. Journal of Biological Chemistry 269 Miyazawa S, Kondo K, Inoue K, Kagamiyama H et al. 21277–21283. 1985b Molecular cloning and nucleotide sequence of the Seedorf U, Raabe M, Ellinghaus P, Kannenberg F, Fobker M, cDNA for rat peroxisomal enoyl-CoA: hydratase-3- Engel T, Denis S, Wouters F, Wirtz KW, Wanders RJ, hydroxyacyl-CoA dehydrogenase bifunctional enzyme. MaedaN&AssmannG1998Defective peroxisomal Journal of Biological Chemistry 260 8905–8910. catabolism of branched fatty acyl coenzyme A in mice Palosaari PM & Hiltunen JK 1990 Peroxisomal bifunctional lacking the sterol carrier protein-2/sterol carrier protein-X protein from rat liver is a trifunctional enzyme possessing gene function. Genes and Development 12 1189–1201.

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access 240    and   · 17â-HSD 4

Sierralta W, Truitt AJ & Jungblut PW 1978 Studies on the oxidation pathway: identification of pristanal as product of involvement of lysosomes in estrogen action. I. Isolation and the decarboxylation of 2-hydroxyphytanoyl-CoA. Biochemical enzymatic properties of pig endometrial lysosomes. Hoppe- and Biophysical Research Communications 237 33–36. Seyler’s Zeitschrift fu¨r Physiologische Chemie 359 517–528. Wanders RJA, Schutgens RBH & Barth PG 1995 Peroxisomal SrivastavaS&Srivastava SP 1991 Effect of di(2- disorders: a review. Journal of Neuropathology and ethylhexyl)phthalate on 17â-hydroxysteroid dehydrogenase Experimental Neurology 54 726–739. in testis of rat. Toxicology Letters 57 235–239. Wanders RJ, Denis S, Wouters F, Wirtz KW & Seedorf U Steinman HM & Hill RL 1975 Bovine liver crotonase (enoyl 1997 Sterol carrier protein-X (SCPx) is a peroxisomal coenzyme A hydratase). Methods in Enzymology 35 136–139. branched-chain beta-ketothiolase specifically reacting with Suzuki Y, Jiang LL, Souri M, Miyazawa S, Fukuda S, Zhang 3-oxo-pristanoyl-CoA: a new, unique role for SCPx in Z, Une M, Shimozawa N, Kondo N, OriiT&HashimotoT branched-chain fatty acid metabolism in peroxisomes. 1997 -3-hydroxyacyl-CoA dehydratase/-3-hydroxyacyl- Biochemical and Biophysical Research Communications 236 CoA dehydrogenase bifunctional protein deficiency: a newly 565–569. identified . American Journal of Human Wirtz KW 1997 Phospholipid transfer proteins revisited. Genetics 61 1153–1162. Biochemical Journal 324 353–360. Swinkels BW, Gould SJ, Bodnar AG, Rachubinski RA & Wu L, Einstein M, Geissler WM, Chan HK, Elliston KO & Subramani S 1991 A novel, cleavable peroxisomal targeting Andersson S 1993 Expression cloning and characterization of signal at the amino-terminus of the rat 3-ketoacyl-CoA human 17 beta-hydroxysteroid dehydrogenase type 2, a thiolase. EMBO Journal 10 3255–3262. microsomal enzyme possessing 20 alpha-hydroxysteroid TremblayY&Beaudoin C 1993 Regulation of 3â- dehydrogenase activity. Journal of Biological Chemistry 268 hydroxysteroid dehydrogenase and 17â-hydroxysteroid 12964–12969. dehydrogenase messenger ribonucleic acid levels by cyclic Yamamoto R, Kallen CB, Babalola GO, Rennert H, Billheimer adenosine 3*,5*-monophosphate and phorbol miristate acetate JT & Strauss JF 1991 Cloning and expression of a cDNA in human choriocarcinoma cells. Molecular Endocrinology 7 encoding human sterol carrier protein 2. Proceedings of the 355–364. National Academy of Sciences of the USA 88 463–467. Tseng L & Gurpide E 1974 Estradiol and 20á- Zhang Y, Dufort I, Soucy P, Labrie F & Luu-The V 1995 dihydroprogesterone dehydrogenase activities in human Cloning and expression of human type V 17â-hydroxysteroid endometrium during the menstrual cycle. Endocrinology 94 dehydrogenase. Proceedings of the 77th Annual Meeting of the 419–425. Endocrine Society 622. Tseng L & Gurpide E 1975 Induction of human endometrial estradiol dehydrogenase by progestins. Endocrinology 97    3 December 1998 825–833. Tseng L & Gurpide E 1979 Stimulation of various 17â-and 20á-hydroxysteroid dehydrogenase activities by progestins in human endometrium. Endocrinology 104 1745–1750. NOTE ADDED IN PROOF TsengL&Mazella J 1981 Kinetic studies of human endometrial hydroxysteroid dehydrogenase. Journal of Steroid Biochemistry 14 437–442. Radiation hybrid mapping by Genethon assigned Verhoeven NM, Schor DS, ten Brink HJ, Wanders RJ & HSD17B4 gene to interval D5S471–D5S393 with a Jakobs C 1997 Resolution of the phytanic acid alpha- physical position of 484.22 cR3000 on .

Journal of Molecular Endocrinology (1999) 22, 227–240

Downloaded from Bioscientifica.com at 10/01/2021 03:29:19PM via free access