<<

d. Biochem. Molec. Biol. Vol. 43, No. 8, pp. 779--804, 1992 09600760/92 $5.00+0.00 Printed in Great Britain. All rights reserved Copyright © 1992 PcrgamonPress Ltd

STEROIDOGENIC : STRUCTURE, FUNCTION, AND ROLE IN REGULATION OF STEROID

ISRAEL HANUKOGLU Department of Hormone Research, Weizmann Imtitute of Science, Rehovot 76100, Israel (Fax: 972-8-344116)

S-mmmT--In the pathways of biosynthesis there are two major types of enzymes: cytochromes P450 and other steroid . This review presents an over- view of the function and expression of both types of enzymes with emphasis on steroidogenic P450s. The final part of the review on regulation of steroidogenesis includes a description of the normal physiological fluctuations in the steroid output of and , and provides an analysis of the relative role of levels in the determination of these fluctu- ations. The repertoire of enzymes expressed in a steroidogenic cell matches the cell's capacity for the biosynthesis of specific . Thus, steroidogenic capacity is regulated mainly by tissue and cell specific expression of enzymes, and not by selective activation or inhibition of enzymes from a larger repertoire. The quantitative capacity of steroidogenic cells for the biosynthesis of specific steroids is determined by the levels of steroidogenlc enzymes. The major physiological variations in enzyme levels, are generally associated with parallel changes in expression. The level of expression of each steroidogenlc enzyme varies in three characteristics: (a) tissue- and cell-specific expression, determined during tissue and cell differentiation; (b) basal expression, in the absence of trophic hormonal stimulation; and (c) hormonal signal regulated expression. Each of these three types of expression probably represent the function- ing of distinct gene regulatory elements. In adult steroidogenic tissues, the levels of most of the ceil- and tissue-specific steroidogenic enzymes depend mainly on trophic hormonal stimulation mediated by a complex network of systems.

OUTLINE 4.1. Enzyme expression as a determinant of steroidogenic capacity 1. Introduction 4.2. Methodological issues 2. Enzymes in the Initial Pathways of Steroido- 4.3. Adrenal cortex genesis 4.4. Testis 2.1. P450scc 4.5. 2.2. 3~-HSD 4.6. Mechanisms of regulation of steroido- 2.3. P450c17 genic enzyme levels 2.4. P450c21 4.7. Intracellular regulators of steroidogcnic 2.5. P450cll and P450c18 enzyme expression 2.6. P450arom 2.7. 17~-HSD 2.8. Enzymes that further metabolize steroids 1. INTRODUCTION 3. Biochemistry and Molecular Biology of Steroidogenic P450 Systems The concentrations of steroid in 3.1. Enzymology of P450 systems blood fluctuate with a specific periodicity, or 3.2. Intracellular targeting of the P450 in response to physiological or pathological system enzymes changes, to regulate diverse processes in the 3.3. Sequence and structural similarities body. In mammals there are three endocrine 4. Role of Enzyme Expression in Regulation of organs that specialize in steroid hormone Steroidogenesis production: adrenal cortex, ovary, and testis. During pregnancy, develops as an additional major source of steroid hormones. Proceedings of the First International Symposium on A Molecular View of Steroid Biosynthesis and , The steroid hormone output of these organs is Jerusalem, Israel, 14-17 October 1991. regulated by specific extracellular hormones and 779 780 ISRAEL HANUKOGLU ._ ii~ii ! iiL il c~ iiii!ili!iiii

¢ ii!~iiiiiiiii ~i~i~ -- 17-OH-Pmgnenolone , DHEA ii ~i~::::

Proou~rone i~i~i~i - 17-OH-Progeaterone ..... , Andromce4ione N~i: FJtrone

Codicosterone Corlisol

18-OH-Cortlcosterone

Aldoeterone J 1 1 1 1 END PRODUCT: Fig. 1. Pathways of initial steps of steroid hormone biosynthesis in the adrenal cortex and the gonadal cells. The arrows mark substrate conversion to product by the enzyme listed in the shaded area. The enzymes in the shaded area with borders (P450sec, P450cl I and P450c18) are located inside the mito- chondria. The other enzymes are located in the . The physiological function of each steroid end product is listed below the frame. The steroids that appear in this figure may be further metabolized within the steroidogenic tissues or in other organs by the enzyme listed in Table 2. DHEA: 3p-hydroxy-5-androsten- 17-one (dehydroepiandosterone). factors that activate a series of intracellular hormonal stimulation but even these high processes that ultimately change the steroid concentrations are not sufficient to supply output. enough steroids to increase blood levels [36, 37]. In our current understanding, steroid hormone This contrasts with the mode of secretion of output depends on the rate of steroid metabol- hormones and neurotransmitters which ism, i.e. biosynthesis and catabolism[l]. The accumulate in intracellular vesicles and are pathways of steroid hormone biosynthesis start secreted in response to specific stimuli. with (Fig. 1). Cholesterol is stored Thus, steroid hormone output is mainly in esterified form inside vesicles. Trophic regulated by events that ultimately affect hormones activate a chain of reactions that lead steroid production through four parameters or to the hydrolysis of cholesterol esters into free processes: cholesterol, and the transport of cholesterol into mitochondria where it is converted to pregnenol- (1) level, determined one by P450scc [2-4]. After this rate-limiting by transcription, stability and translation step, subsequent biosynthetic steps proceed of the mRNAs encoding the enzymes. with the flow of substrates through the enzyme (2) Steroidogenic enzyme activity, determined systems located in the endoplasmic reticulum by the conditions of the intracellular and mitochondria. Steroid hormones are hydro- milieu, availability, or the post- phobic molecules that can penetrate biological translational modification of the enzymes. membranes, and flow into the blood stream (3) Substrate availability, generally deter- after their synthesis without being stored in mined by cholesterol mobilization and intracellular vesicles. An increase in the blood transport to the mitochondrial P450scc levels of steroid hormones is dependent on the which catalyzes the first step in the continual synthesis and secretion of steroids. pathways of steroid biosynthesis. Tissue levels of steroids rise transiently after (4) Tissue growth, determined by cell division Steroidogenic enzymes 781

Table 1. Enzymes that catalyze the initial reaction in the pathways of steroid hormone biosynthesis (see Fig. I) Nomenclature Enzyme Gene Reaction Reviews P450 type P450~ CYP l lA Cholesterol side chain cleavage 2-4, (5) (3 successive monooxygenations) P450cl I CYPIIBI C-I 1 4, 6, (7-9) C- 18 hydroxylation P450cl8 CYPIIB2 C- 18 hydroxylation 6, (7-10) C-I 8 oxidation P450c17 CYP I 7 17,, -Hydroxylation 11, (I 2) C 17---C20 bond cleavage P450c21 CYP21 C-21 hydroxylation ! 1, (13, 14) P450arom CYPI9 Steroid ring-A aromatization 15, 16 (3 successive monooxygenations) (dehydrogenase) type 3#-HSD HSD3B 3p-Hydroxysteroid dehydrogenation 17 (5) 5-Ene-4-ene isomerization 17p-HSD 17/I-Hydroxysteroid dehydrogenation 18-20, (21) 17-Keto-steroid reduction The references in parentheses include description of inborn deficiencies of enzyme. The table includes at most only two reactions for each enzyme, based on the highest amount of products observed in in vitro incubations with the enzyme. All the enzymes listed catalyze additional reactions with different stereoselectivities, but at lower yields than the major reactions listed above.

and multipScation, as in the expression of both types of enzymes and then formation. expounds on the structure and regulation of steroidogenic P450s. Steroid dehydrogenases Several of these processes may operate con- [17], and P450s involved in the biosynthesis of currently to determine the final steroid hormone and are reviewed in other output. For example, growth of corpus luteum sections of this issue [44]. is accompanied by changes in the expression The final part of this review on the regulation of steroidogenic enzymes (Section 4.5). Trophic of steroidogenesis includes a description of the hormonesmay also activate several of these four normal physiological fluctuations in the steroid processes simultaneously. Whereas, cholesterol output of adrenal cortex and gonads and supply can be increased within minutes, the provides an analysis of the relative role of the induction of enzyme synthesis may require enzyme levels in the determination of these fluc- many hours. Hence, these actions of stimulatory tuations (for reviews on placental steroidogenesis hormones have been referred to as 'rapid' vs see Refs [45, 46]). 'delayed' or 'trophic'. However, the period of initiation of these responses may overlap. For 2. ENZYMES IN THE INITIAL PATHWAYS example, brief (30 min) stimulation of adreno- OF STEROIDOGENESIS cortical cells with ACTH leads to both a rapid increase in steroid secretion and a delayed The initial steps of steroid biosynthesis in increase in enzyme synthesis that peaks 36 h steroidogenic tissues are shown in Fig. 1. later [38]. Activity and expression of the enzymes in Among the 4 processes mentioned, this review different tissues are listed in Tables 1-3. During concentrates on the role of steroidogenic the past decade, cDNAs for nearly all the enzyme expression and activity as a determinant of steroidogenic capacity. Studies on the tran- Table 2. A partial list of enzymm that further metabolize steroids scriptional regulatory sequences in that Itefe~nc~ P450 type encode the enzymes are the subject of other P450s that hydroxylate steroids at positions 22-29 reviews in this issue[16,39-42]. Cholesterol 2% 2p, 6~, 7,,, 15,,, 15p, and 16~ Oxidoreductaae (dehydrogenase) type transport is also reviewed in another section of 3,, -OH-steroid dehydrogenase 30 this issue [2, 43]. 5~-Rednctase 31 5F-Rednctase In the pathways of steroid hormone biosyn- I 1# -OH.~teroid dehydrogenaae 8 thesis there are two major types of enzymes: 20~-OH-steroid dehydrogenue 32 20# -OH-steroid dehydrogenase cytochromes P450 and other steroid oxido- Conjugating enzyme type reductases (Fig. 1 and Table 1). Initially this Sulfotrnosfera~ 33 review presents an overview of the function and UDP glncuronosyltrnn~erase 34, 35 782 ISRAEL HANUKOGLU

Table 3. Specificity of expression of steroidogenic enzymes in different ceil types in the adrenal cortex, ovary and testis Adrenal cortex Ovary Testis Luteinized Luteinized Granulosa Granulosa thcca granulosa Leydig Zona Zona Theca (preantral (preovulatory (corpus (corpus (interstitial glomerulosa faseiculata interna follicle) follicle) luteum) luteum) tissue) P450-~c + + + -- + ++" ++ + 3p-HSD + + + + + ++ ++ + P450cl 1 -- + ...... P450cl 8 + P450cl 7 _{_b + _[_ .{_ P450c2 I + + ...... P450arom .... + -- +c -- 17p-HSD ? + + .... + Major steroid Miueralo- Glucocorticoid Androgen Estrogen Progestins Prosesterone Androgen product corticoid Androgen progesterone Androgen Estrogen • + +, indicates much higher levels only relative to ovarian granulosa ceils and not other ceils. bNot expressed in rat, hamster and rabbit adrenals. CExpression dependent on species. major steroidogenic enzymes have been isolated a single gene for each enzyme must carry all (Table 4). One of the most important findings the signals for its specific regulation in different that emerge from molecular studies is that tissues and cells. A second most important find- almost every steroidogenie enzyme is encoded ing is that some of the enzymes, e.g. P450c17, by only one gene (Table 4). For key enzymes, P450c18, catalyze more than one step in the e.g. P450see, different genes that are specifically pathway (Fig. 1). Therefore, it is recommended expressed in different tissues do not exist. Thus, that the enzymes be referred to with a single

Table 4. Characteristics of the genes and mRNAs of cytochromes P450 and their electron transfer Gene mRNA Gene length length length Species (n) Chromosome (kb) (kb) (aa) (preseq.)" References Mitochondrial Adrenodoxin reductase Man I 17 12 2 459 (491) 47-51 Cow I - 6-12 2 460 (492) 47, 52, 53 Andrenodoxin Man 2 (2) b 11 (20, 21) >20 I, 1.4, 1.7 124 (184) 51, 54, 55 Cow I - > 27 I, 1.4, 1.7 128 (186) 56-59 P4508¢c Man 1 15 >20 2 482(521) 51,60-62 Cow 1 - - 2 481 (520) 63-65 Pig .... 481 (520) 66 Rat 1 - - 2 490 (526) 67, 68 P450cl I Man 1 8 - - 479 (503) 7, 8, 69, 70 Cow 2 (2) - - 1.8, 4.1 479 (503) 71-74 Rat .... 475 (499) 6, 75 Mouse I 15 7 - (500) 76 P450cl 8 Man I 8 - - 479 (503) 7, 8, 69, 77 Rat - (7) - - - 476 (510) 6, 78 Mouse 1 15 7 2.6 (500) 76

Microsomal P 4 50red. Human 1 - - - 676 79, 80 Porcine .... 678 81 Rabbit - - - 2.4 679 82 Rat 1 - 20 2.7 678 79, 83-85 Mouse I 6 ------79 P450cl7 Man I 10 6.6 1.9 508 51, 86--88 Cow 1 - 6.6 1.9 509 89, 90 Rat 1 - - 1.9 507 91, 92 Mouse 1 19 - - 507 93 Chicken - - - 1.9 508 94 P450c21 Man 1 (1) 6 3.3 2 494-495 13, 95, 96 Cow 1 (1) - 3.4 2.2, 2.4 496 97-101 Mouse I (I) 17 3.1 2.2 487 102, 103 P450arom Man I 15 > 70 2.9, 3.4 503 16, 104-108 Rat .... 508 109 Mouse 1 9 - 2.1, 2.5 503 I i0 Chicken - - - 4 507 I ! 1 Trout - - - 2.6 522 ! 12 "The presequence of the protein includes amino terminal extension signal peptide. el'he number of l~endogeues are listed in parentheses. Steroidogenic enzymes 783

Table 5. The concentration of mitochondrial P450scc in steroido- P450 nomenclature and not with a commonly genie tissues and brain used "hydroxylase" name that would require Concentration the use of two or more hydroxylase names for Tissue (prnol/ms protein) ° Reference a single enzyme molecule [25]. Adrenal cortex 400 121 Corpus luteum 80-400 121,122 The first two major steps in the biosynthesis Ovary <5 121,122 of steroids are common to all steroidogenic Placenta < 50 123 Testis Leydig cells - 119 organs: (1) conversion of cholesterol to preg- Brain < 10 124 nenolone by P450scc inside the mitochondria; 'The values shown are per total tissue pro~'in. The concentrations and (2) conversion of pregnenolone to pro- for placenta and brain were calculated assuming that the mito- gesterone by 3fl-hydroxysteroid dehydrogenase cbondrial fraction represents 20% of the total cellular protein. (3fl-HSD) in the endoplasmic reticulum (Fig. 1). The following steps of steroidogenesis are genomes there are at least 2-3 homologous catalyzed by enzymes expressed only in some genes encoding 3fl-HSDs that share 80-94% steroidogenic cell types (Table 3). The cell- sequence identity within each species [17]. Two specific expression of these enzymes determines types of 3fl-HSDs from human and rat can the capacity of each cell to synthesize different use either pregnenolone, 17-OH-pregnenolone steroid hormones (Table 3). The major functions or dehydroepiandosterone (DHEA) as substrates of these enzymes and their sites of expression [17]. The different types have different Km values are listed below. for the same substrates. However, each type of enzyme can use pregnenolone or DHEA as sub- 2.1. P450scc strates with similar K,, values [17]. Rat type I This enzyme catalyzes the first and rate- 3fl-HSD also shows 17fl-HSD activity with limiting step in the biosynthesis of steroid 5u-androstane steroids but not with , hormones [2-4, 113, 114]. It converts cholesterol , , or [125]. to pregnenolone in three successive monooxy- In the human adrenal cortex and gonads, but genations ( at C-22, followed by not placenta, only type II 3~-HSD is expressed, C-20, and finally cleavage of the C-20,22 bond). whereas in the rat both type I and II are found Hydroxylated intermediates of cholesterol bind in these tissues [17]. Immunohistochemical very tightly to P450scc and do not show studies reveal localization of 3fl-HSD in the significant dissociation from the enzyme [4]. In same steroidogenic cells as P450scc in all three contrast, the final product pregnenolone has a zones of the adrenal cortex, in the interstitial dissociation constant 40 to 600-fold higher than Leydig cells of the testis, but not in seminiferous those of intermediates, facilitating its release tubules, in the theca interna of the ovary and from the enzyme [4]. Purified P450scc can effici- in corpora lutea [126]. Immunostaining failed to ently catalyze side chain cleavage of cholesterol detect 3fl-HSD in rat granulosa cells [126] even sulfate, and 6fl-hydroxylation of deoxycortico- though these cells display high 3fl-HSD activity sterone [115, 116]. Yet, it is not known whether converting pregnenolone to progesterone even these reactions represent a major activity in vivo. before the induction of P450scc [127]. In con- P450scc is expressed in all three zones of the trast to stcroidogenic P450s, 3fl-HSD activity is adrenal cortex [117, 118]. In the testis it is found present in a wide range of tissues [17]. in the steroidogenic Leydig cells[ll9]. In the ovary it is expressed in the theca interna; its 2.3. P450c17 expression in the granulosa cells depends on the This enzyme catalyzes two key reactions: stage of growth of the follicle as discussed in (a) 17u-hydroxylation of C21 steroids; and (b) Section 4.5. In addition to steroidogenic tissues cleavage of the C17---C20 bond of C21 steroids. the expression of P450scc has been detected in The 170t-hydroxylation is a required step in the brain [120], but at levels that are more than biosynthesis, whereas the C17---C20 an order of magnitude lower (Table 5). bond side chain cleavage is essential for the biosynthesis of (Fig. 1). Genetic 2.2. 3fl-HSD deficiencies affecting either one or both activities This enzyme has two major catalytic activities have been characterized [12]. which in concert convert 3fl-hydroxy-5-ene Immunohistochemical analysis shows that steroids into 3-keto-4-ene (Table 1). In contrast P450c17 is present in the zona fasciculata and to steroidogenic P450s each of which is encoded reticularis but not in zona glomerulosa of the by a single gene, in the human, rat and mouse porcine adrenal [128]. This is consistent with the 784 ISRAEL HANUKOGLU role of zona glomerulosa as the site of mineralo- specificity of P450c17 determines whether corticoid biosynthesis where 17~-hydroxylation androgen biosynthesis proceeds mainly through is not required (Fig. 1). P450c17 is not expressed pregnenolone or progesterone as shown in Fig. 1. in the rat adrenal cortex; consequently cortico- The formation of androgen may be catalyzed sterone, and not cortisol, is the major glucocor- without the 17~-hydroxylated substrate leaving ticoid in this species and in some other rodents the enzyme [141]. [129]. However, P450c17 is expressed in guinea pig adrenal cortex [130-134]. 2.4. P450c21 In the ovary, P450c17 is expressed in theca This enzyme catalyzes an essential step in interna cells[135, 136]. Antibody or cDNA the synthesis of gluco- and probes show very low expression of P450c17 (Fig. 1). Congenital adrenal hyperplasia which in granulosa cells of humans and rats[135- results from an inborn deficiency of this enzyme 137]. Thus, theca interna cells can synthesize is a common genetic disorder [5, 13, 14]. Defici- androgens, but granulosa cells which produce ency in the P450c21 hydroxylation step, channels are dependent on androgen precursor the steroid biosynthetic pathway in the direction supply from theca interna[138]. This process of androgen production, resulting in gluco- is called the two cell hypothesis of follicular and mineralocorticoid deficiency and excessive estrogen production [138]. virilization [5, 14]. The fact that P450cl 7 catalyzes two reactions, P450c21 is expressed in all three zones of the poses a conundrum: What determines the relative adrenal cortex [146]. Apart from the adrenal cor- flow of steroids through glucocorticoid and tex, P450s with steroid 21-hydroxylation activity androgen pathways (see Fig. 1)? Adrenal cortex have been found in other tissues, e.g. [147]. capacity for androgen synthesis changes during However, these P450s show little similarity [139]. These pathways are probably to the steroidogenic P450c21 and represent developmentally regulated by changes in the products of other genes [25, 147]. Immunohisto- function of the zona reticularis. In Leydig chemical studies using anti-P450c21 antibody cells, in the absence of P450c21 there is no revealed a cross reacting protein only in the alternative pathway but to androgen production distal tubules of the bovine , which is the (see Fig. 1). site of the mineralocorticoid action in the kidney The C17--C20 bond cleavage activity of [146]. However, it is not known whether this P450c17 depends on the concentration of elec- protein is the product of the same gene that tron transfer protein P450 reductase, and it can encodes the adrenal P450c21. Examination of be increased to the level of the 17~-hydroxyl- the kidney by in situ hybridization using a ation activity [11]. Thus, changing levels of this specific P450c21 probe could resolve this reductase or cytochrome b5 may affect the ratio question. of the two activities [11,140]. Different steroido- genic tissues appear to have different levels 2.5. P450cli and P450c18 of these proteins[l 1]. The two activities of These two enzymes are also uniquely expressed P450c17 are also strongly dependent on steroid in the adrenal cortex [6, 148]. In the human and concentrations [141,142]. Thus, the intracellular rat adrenal, P450cl 1 is expressed in the zona concentrations of substrates and products prob- fasciculata which specializes in glucocorticoid ably also play a role in regulating the relative production under mainly ACTH regulation. rates of these activities [141,142]. Whereas P450c18 is expressed in the zona The substrate specificity of P450c17 varies glomerulosa and is regulated by the - with species[129]. P450c17 from all species angiotensin system [6-8, 149-151]. The human examined can hydroxylate C-17 of both preg- and rat P450cl 8 can catalyze form- nenolone and progesterone, although the Km for ation, but P450cll cannot [6-8]. Two bovine these two substrates can differ by 10-fold in isozymes of P450cl I expressed in COS-7 cells some species [11, 91,143-145]. Whereas, the rat can catalyze both and aldosterone P450cl 7 can convert both 17-OH-pregnenolone production[152]. However, it is not known and 17-OH-progesterone into DHEA and andro- whether these enzymes function in both zones, stenedione, respectively [91,143], the human and or whether there is a bovine P450c18 gene that bovine P450c17 can cleave the C17--C20 bond is specifically expressed in zona glomerulosa. of 17-OH-pregnenolone but not of 17-OH- With 11-deoxy substrates, both P450cl 1 and progesterone [91,144, 145]. Thus, the substrate P450c18 also catalyze 19-hydroxylation but at Steroidogenic enzymes 785 less than 10% of 1 lp-hydroxylation activity [7]. disorder, but is observed frequently in an inbred P450cl 1 can also hydroxylate androstenedione Arab population[21]. In affected individuals resulting in the formation of 11/~-hydroxylated androstenedione to testosterone conversion is androgens [e.g. 134]. Steroid 11/~-hydroxylation markedly impaired [21]. The cloning of the gene activity has been detected in the gonads of some that encodes the androgenic 17/~-HSD may be species [153], but it is not known whether this useful in elucidating the molecular defect in this reflects a low expression of P450cl 1 in these disease. tissues or the activity of another enzyme. Immunohistochemical studies using an antibody against a testicular 17//-HSD indicate 2.6. P450arom that the enzyme is expressed in the testis in P450arom catalyzes the conversion of Leydig cells of the interstitial tissue, and in the testosterone into the 17/~-estradiol (Fig. 1 and ovary, in the theca interna, but not in granulosa Table 1). In the ovary P450arom is expressed ceils or corpus luteum [157]. This is consistent in granulosa cells which is the major site of with androgen synthesis in testicular Leydig estrogen production in females [136, 154] (see cells and in ovarian theca interna. Both andro- Section 4.5). However, this enzyme is widely genic and estrogenic 17p-HSD activities have expressed in many tissues besides gonads, e.g. been observed in a wide range of tissues [20]. adipocytes, breast, central nervous system, These activities probably do not represent the skin and placenta[15, 16]. The gene encoding androgenic isozyme. In testicular 17I/-HSD P450arom is the longest amongst steroidogenic deficiency, 17/~-HSD activity is defective only P450 genes (Table 4). This gene is also unique in the testis but appears normal in other among P450 genes in having alternative tissues [21], thus supporting other findings that promoters that are utilized in a tissue-specific there are multiple isozymes with 17f/-HSD manner [16]. activity [18-20]. 2.7. 17[$-HSD 2.8. Enzymes that further metabolize steroids This enzyme is also referred as 17-keto- The enzymes noted above catalyze the initial . It catalyzes the reversible steps in steroidogenesis. The end products of conversion of the 17-keto and 17/~-hydroxy these reactions can be further metabolized by groups in androgens and estrogens, including other enzymes (Table 2) within the cell of androstenedione, DHEA, and 17/~-estradiol. production or after transport to cells in the The direction of the reaction depends on the sub- same or other organs. Some properties of these strate and cofactor [18-20]. For the conversion enzymes are outlined below: between androstenedione and testosterone, the porcine testicular 17fl-HSD prefers NADPH (1) Steroid metabolism by some of these rather than NADH (Kin = 11 vs 177 #M) [155]. enzymes may result in the formation of There are multiple 17fl-HSD isozymes with a steroid product with potent biological androgen or estrogen specificity [18-20]. The iso- activity, as in the case of conversion of zyme from the porcine testis also displays 200t- testosterone to 5~- by HSD activity at about a tenth of the 17fl-HSD 5~t-reductase, or inactivation of the activity [155]. steroid, as in the case of liver hydroxyl- A 17fl-HSD has been purified from placenta, ation and conjugation of steroids, or and a eDNA and two in tandem homologous inactivation of cortisol by 1 lfl-HSD (see genes have been cloned and sequenced [19, 156]. refs in Table 2). This enzyme catalyzes the interconversion of (2) In contrast to the initial steroidogenic 17/~-estradiol and estrone, and can also use enzymes which are encoded by a single androgen substrates [19]. Its Km for 17/~-estradiol gene (Table 4), the cloning of cDNAs for is 10#M vs 250#M for testosterone[19]. some of these enzymes revealed large Thus, this enzyme is specific for estrogens and families of genes with many members (see not androgens. Its cDNA hybridizes relatively refs in Table 2). The enzymes encoded by weakly to a few bands on a testis RNA blot these genes have similar sequences, but [156]. One of these may represent a homologous different substrate specificities. mRNA that encodes a different enzyme with (3) While most steroidogenic enzymes have specificity for androgens rather than estrogens. strict substrate specificities, the enzymes Testicular 17/~-HSD deficiency is a rare genetic that further metabolize steroids generally 786 ISRAEL HANUKOGLU

have broader substrate specificities. For This reaction consumes two electrons, a H +, example, 30t-HSD and 20~-HSD can and 02, one atom of which is incorporated into metabolize both C19 (androstane) and the substrate, while the second is reduced to C21 (pregnane) steroids. Similarly, some H20 (hence mono-oxygenation). The electrons conjugating enzymes can accommodate a are transferred from NADPH to cytochrome broad range of steroid structures. The P450 by specific electron cartier proteins. In the stereoselectivities of some enzymes may mitochondrial P450 systems this function is also be broader: different P450s can performed by two proteins: adrenodoxin reduct- hydroxylate a steroid at several positions ase, which is an FAD containing , [28] and 20at-HSD also shows 3ct-HSD and adrenodoxin, which is a ferredoxin type activity [32]. iron-sulfur protein [3, 4, 47, 113, 114, 160]. The (4) Whereas the expression of most enzymes microsomal P450 systems are dependent on in the initial pathways (Fig. 1) is limited to P450 reductase which is a flavoprotein with steroidogenic tissues, enzymes of further FAD and FMN as cofactors [22, 23, 47, 161]. In metabolism (Table 2) are expressed in a a catalytic cycle, the mitochondrial P450 accepts broad range of tissues besides steroido- one electron at a time from adrenodoxin, and genic organs. For example, 3ct-HSD, and the microsomal P450 from the FMN of P450 5~t-reductase activities are present in reductase. Thus, the cofactor FMN fulfills a role many peripheral tissues as well as all areas similar to that of adrenodoxin in the mitochon- of the central nervous system including drial P450 systems as a single electron acceptor white matter [158, 159]. and donator[161] (Fig. 2). Some microsomal P450s can also accept the second electron from cytochrome bs [11, 22, 140]. 3. BIOCHEMISTRY AND MOLECULAR BIOLOGY All eukaryotic P450s are membrane associ- OF STEROIDOGENIC P450 SYSTEMS ated enzymes. The amino termini of both the microsomal P450s and P450 reductase invari- 3.1. Enzymology of P450 systems ably contain a 20-30 residue long highly hydro- As noted above, most of the reactions in the phobic segment which anchors these enzymes to pathways of steroid biosynthesis are catalyzed the of the endoplasmic reticulum, by P450 type enzymes. P450s are found in while their main body remains in the cytoplasmic nearly all tissues of the vertebrates where they side[162, 163,163a] (Fig. 2). Mitochondrial catalyze different metabolic or biosynthetic P450s are located on the matrix side of the inner reactions generally involving small hydrophobic mitochondrial membrane [117, 118, 164]. The molecules. The steroidogenic P450s resemble mitochondrial P450s behave as highly hydro- other P450s in many biochemical properties. phobic proteins, but contrary to the microsomal All cytochromes P450 are b type cytochromes P450s they lack a hydrophobic amino terminal wherein the environment of the heme forms the sequence that could be predicted as a membrane of the enzyme that binds the substrate spanning segment [71,165, 166]. Thus, the and 02[22, 23]. CO competes with 02 for region that anchors the mitochondrial P450s binding to this site and thus can inhibit to the membrane remains unidentified. Both enzymatic activity. The name P450 derives from adrenodoxin reductase and adrenodoxin are the distinct absorption maximum at 450 nm easily solubilized proteins, and do not contain a of the reduced and CO bound form of the highly hydrophobic segment[47]. However, enzyme. immunocytochemical studies show that these The reactions catalyzed by steroidogenic proteins are also associated with the inner mito- P450s are either a single hydroxylation (mono- chondrial membrane [167], most likely mainly oxygenation) at a specific position on the steroid by ionic interactions. molecule, or a series of consecutive monooxy- Generally the reductase component of P450 genations which result in C---C bond cleavage systems is present at much lower concentrations or aromatization of the steroid ring A (Table 1). than the P450s inside cells [121,168]. This low Each P450-catalyzed hydroxylation has the molar ratio of reductase to P450, and kinetic following stoichiometry: studies of enzyme--enzyme interactions indicate that the P450 system enzymes do not form Steroid-H + NADPH + H ÷ + 02 static complexes on the membrane and are Steroid-OH + NADP ÷ + n20 independently mobile [22, 113, 160]. Steroidogenic enzymes 787

Adr. reductase Adrenodoxin P450 NAF-

NAI

Inner mitochondrial membrane

P450-Reductase P450

Endoplasmic reticulum membrane Fig. 2. Models for the membrane organization of the mitochondrial and microsomal P450 systems. The mitochondrial system faces the matrix (inner) side of the membrane. The microsomal system enzymes face the cytosol and both are anchored to the membrane by hydrophobic amino terminal domains. The membrane binding region of the mitochondrial P450s is not known and is depicted in analogy to the microsomal P450s.

3.2. Intracellular targeting of the P450 system drial precursor proteins with the mitochondrial enzymes membrane may be mediated by the polar heads of cardiolipin, a phospholipid that is specifically The mitochondrial P450s, adrenodoxin present in the inner mitochondrial membrane reductase and adrenodoxin are synthesized in [177]. The bovine mitochondrial P450s can be precursor forms with an amino terminal exten- imported only into mitochondria isolated from sion of 30-60 residues that is absent in the steroidogenic tissues, whereas the precursor of mature enzymes (Table 4). These amino terminal adrenodoxin which has a wider tissue distri- signal are necessary for the specific entry bution, does not show this tissue specificity of the enzymes into mitochondria[169-171], [175-178]. The sequence or structural features and are cleaved by proteases upon entry of the responsible for this tissue specificity of transfer precursor into the mitochondria[172-175]. In have not yet been identified. bovine adrenal cortex, P450scc and adrenodoxin The role of the mitochondrial targeting are processed by two distinct proteases [173, 175]. signals has also been examined using expression The sequences of the extensions of these proteins plasmids containing cDNAs with truncated share little or no similarity, are not hydrophobic, or modified signal peptide sequences. Mature and invariably contain several basic residues bovine adrenodoxin reductase and adrenodoxin (Arg and Lys) which are essential for the transfer sequences expressed in yeast (heterologous of at least some of these proteins into mitochon- expression) were found in the cytosolic fraction dria [169-171]. In these respects, these proteins because of the lack of the mitochondrial signal resemble some other mitochondrial proteins peptide[179]. Whereas, with a spliced yeast [176]. The specific association of the mitochon- mitochondrial cytochrome c oxidase subunit 788 ISRAEL HANUKOGLU signal peptide sequence, the expressed proteins but evolutionarily related families of were directed into the mitochondrial fraction genes. [179]. (6) The cDNA sequences of most of the Unlike the mitochondrial P450s, the micro- human and bovine steroidogenic P450 somal P450s and P450 reductase do not have system enzymes have been determined an extension peptide or basic residues in the (Table 4). The homologous enzymes in amino termini (Table 4). The hydrophobic these two species share >70% sequence amino terminus of the microsomal P450s deter- similarity. mines the membrane entry and association of (7) The flavoprotein reductase components these proteins [162, 163a]. Wild type P450 re- of the mitochondrial and microsomal ductase expressed in yeast is normally incorpor- P450 systems show no sequence similarity ated into the microsomes[180]. However, and belong to different families of truncation of its hydrophobic amino terminus oxidoreductases [47, 52]. produces an enzyme that remains soluble in the (8) The analysis of the sequence of the cytosolic fraction [180]. Thus, this segment ap- mitochondrial adrenodoxin reductase led pears to be the only segment that anchors the to the definition of a new large class of protein to the microsomal membrane. oxidoreductases that share an NADP consensus sequence [47, 52]. 3.3. Sequence and structural similarities These analyses also led to the discovery The biochemical similarities between cyto- of a sequence difference between NAD chromes P450 noted above indicated some and NADP binding sites [52, 182]. degree of structural similarity between these enzymes. The precise sequence similarities were Determination of the 3-D structure of eukary- established after the sequencing of the cDNAs otic P450s has been hindered by the difficulties for each of these enzymes. Comparison of inherent in the crystallization of hydrophobic the sequences of these enzymes revealed the membrane bound proteins. However, the crystal following relationships [25]: structure of a bacterial P450BM-3 which shares significant sequence homology with eukaryotic (1) The microsomal steroidogenic P450s microsomal P450s has been determined[183]. share <40% sequence similarity with This structure can serve as a model to under- each other. Thus each P450 represents a stand the structure of other P450s. The 3-D distinct family with generally a single structure of P450cam from Pseudomonas putida member. In contrast, the liver microsomal was elucidated earlier [184]. Although the struc- P450s, which are involved in the metab- tures of P450BM-3 and P450cam share similar olism of endo- and xenobiotics, belong to structural domains, the only significant sequence large gene families which include many similarity between P450cam and the eukaryotic members with > 50% sequence similarity P450s is observed around the heme binding [25]. region. There were many attempts to align and (2) The three mitochondrial P450s, namely match the full sequence of P450cam with eukary- P450scc, P450cll and P450c18, share otic P450 sequences. However, the elucidation sequence similarity along their entire of the structure of P450BM-3 showed that these length [7, 8, 71]. previous alignments did not correctly match (3) The mitochondrial and microsomal P450s different structural domains of the molecules show sequence similarity only around a (J. A. Peterson and S. S. Boddupalli, personal limited region in the heme binding seg- communication). ment close to the carboxy termini of the Currently the structure of many P450s that enzymes [181]. metabolize steroids is being investigated by (4) The microsomal P450¢17 and P450¢21 expressing modified cDNAs, chimeric cDNAs share sequence similarity in their amino that include parts of different P450s [e.g. 185], terminal regions and carboxy terminal and fused cDNAs that include both P450 and halves. reductase [e.g. 186], in bacteria and yeasts. (5) P450c17 and P450c21 share <30% over- These studies are defining the role of specific all sequence identity with the liver micro- regions and residues involved in substrate bind- somal P450s. Thus, the liver microsomal ing, enzymatic activity and interaction with and steroidogenic P450s represent distinct membrane and other molecules. Some of these Steroidogenic enzymes 789 are reviewed in this issue [24, 26]. The expression adult life. Thus, while the levels of steroids of modified P450s is also being used to examine secreted from these two organs show temporal the deleterious effects of genetic mutations on fluctuations, the major types of steroids do not the enzymatic activity of P450s [187]. change (Section 4.3). In contrast, in ovarian follicles the repertoire of enzymes in granulosa cells changes during the final stages of follicle 4. ROLE OF ENZYME EXPRESSION IN REGULATION OF STEROIDOGENESIS development (Table 3). Consequently, the ovarian steroid output shows persistent major Early studies based on measurements of changes in both the major types and the levels enzyme activities indicated that some changes in of the steroids secreted during the female blood levels of steroids may result from changes reproductive cycle. in the levels of specific steroidogenic enzymes Rapid hourly fluctuations in steroid output ([129] for a historical view see Ref. [188]). cannot reflect changes in enzyme levels, and However, assays of steroid levels in blood or probably generally result from regulation of in vitro steroid conversion in tissue homogenates substrate cholesterol transport. The levels of may reflect many different effects other than steroidogenic enzymes, like most other proteins, enzyme levels. The purification of steroidogenic are determined by the balance of de novo syn- enzymes, production of antibodies and isolation thesis and normal metabolic degradation. For of cDNAs provided refined tools to examine the steroidogenic P450s both of these processes are role of steroidogenic enzymes in regulation of slow, P450s are highly stable proteins. In vitro steroidogenesis. studies indicate a half life of 24-42h[189]. Recent studies in this field have been guided In vivo studies indicate an even longer half life generally by the following questions: (a) What is of several days [190, 191]. The mRNAs encod- the tissue and cell distribution of steroidogenic ing P450scc, P450c11, P450c17, and P450c21 enzymes? (b) Is there a relationship between the are also stable with half lives ranging from 5 to blood levels of steroids and the tissue levels of 30 h [192]. steroidogenic enzymes? (c) Which extraceUular factors and hormones regulate the levels of 4.2. Methodological issues steroidogenic enzymes? (d) What chains of Histochemical techniques are the methods of molecular events are activated by hormones and choice to determine tissue and cellular localiz- other extracellular factors to ultimately change ation of enzymes using anti-enzyme antibodies, steroidogenic enzyme levels? (e) Are the changes or eDNA probes for in-situ hybridization [e.g. in the levels of the enzymes a result of changes 146]. Histochemical techniques can also be used in the transcription, stability or translation of to investigate changes in enzyme levels for a the mRNAs encoding the enzymes? (f) What qualitative assessment. However, quantitation are the gene sequences responsible for tissue of enzyme or mRNA level generally requires the and hormone specific regulation of the genes use of protein or RNA blots which are probed encoding the enzymes? using antibody or eDNA probes, respectively Current research on questions (a-e) is [e.g. 38, 121]. These methods can be applied to summarized below, citing mainly studies that tissue samples directly. Yet, as the investigations utilized antibody and eDNA probes to measure proceed deeper into the molecular level of regu- enzyme and mRNA levels. lation, they require the use of in vitro systems of cultured cells. 4.1. Enzyme expression as a determinant of Although culture conditions represent a steroidogenic capacity drastic departure from in vivo conditions, Recent research established that the repertoire steroidogenic cells in culture maintain many of enzymes expressed in a steroidogenic cell of their physiological responses. Yet, to avoid matches exactly the cell's capacity for the bio- detachment from the physiology of steroid hor- synthesis of specific steroids (Table 3). Thus, mones, in the common use of in vitro systems it steroidogenic capacity is regulated mainly by is important to consider the following questions: tissue- and cell-specific expression of enzymes, (a) Do the culture conditions inhibit or modify and not by selective activation or inhibition of normal physiological responses? (Factors like enzymes from a larger repertoire. substratum for cell growth[193], cell density In adrenocortical and testicular tissue, the [194, 195], medium and atmosphere composition repertoire of enzymes remains constant during [121,196] have been shown to affect the 790 ISRAEL HANUKOGLU responses of steroidogenic cells.) (b) Does the enzymes. Blood glucocorticoid levels are elevated blood or local level of the examined factor show many fold in certain disease states [204, 205], changes that parallel the physiological variation while adrenal androgen levels may be suppressed in the steroid levels? (c) Is the concentration of [205]. Moreover, during acute illness, the the factor within physiological or pharmaco- responsiveness of the adrenal cortex to ACTH logical range? (d) Do the in vitro changes in stimulation increases, indicating enhanced steroidogenic capacity and enzyme levels reflect steroidogenic capacity probably due to increased physiological changes in steroid and enzyme steroidogenic enzyme levels (Hanukoglu A., levels? Fried D. and Hanukoglu I., unpublished observ- ations). ACTH treatment increases the activities 4.3. Adrenal cortex of certain adrenal steroidogenic enzymes in The blood levels of the major steroid products rodents [133, 206], and changes the cellular fine of all three zones of adrenal cortex, vary in morphology [207]. Extensive ACTH exposure a circadian pattern in response to trophic initially stimulates cellular hypertrophy of hormones specific for each tissue[197-200]. adrenocortical cells without causing cell prolifer- Steroidogenic enzyme levels cannot vary drastic- ation [208]. Thus, elevations in blood gluco- ally within 12 h (see Section 4.1). Thus, circadian corticoid levels during acute disease probably variations cannot result from changes in the do not represent adrenal tissue growth. levels of the enzymes, and most probably reflect Plasma levels of do not differ hormonal stimulation of steroid biosynthesis via between males and females[139]. In bovine increased cholesterol transport to mitochondria. adrenal cortex, the levels of P450scc, P450cl 1, This assumption is supported by the observations and their electron transfer proteins are similar in that in rats the diurnal increase in plasma different animals and show no sex difference corticosterone is associated with a diurnal [121]. In guinea pig adrenal cortex the levels of decrease in adrenal cholesteryl esters that serve P450c17 and P450c21 are also similar in both as the precursor of adrenal steroids [200]. In sexes[131]. In inbred mice, strain differences man, the patterns of circadian variation of some have been observed in the levels of steroidogenic adrenal androgens are different from that of enzymes in both adrenal cortex and testis [209]. cortisol, indicating that these may be regulated Some P450s in the guinea pig adrenal cortex mainly by factors different from ACTH [199]. that are immunologically related to the liver The blood levels of adrenocortical steroids P450s, and 5~t-reductase in the rat adrenal rise rapidly in response to conditions that cortex, show sexual dimorphism and are affected stimulate ACTH secretion [201]. After ACTH by sex steroids [131, 210]. These findings indicate injection to human subjects, both plasma cortisol that sex steroids do not have a major influence and aldosterone levels increase several fold on the initial steroidogenic pathway of the within 1 h [139, 202]. However, the plasma level adrenal cortex, but determine later steps of of ACTH, after injection, highly correlates with steroid metabolism in these species. Certain cortisol, but not aldosterone levels, indicating steroid metabolizing enzymes in the liver are that while cortisol secretion is mainly determined also expressed in a sexually dimorphic pattern by ACTH, aldosterone secretion is affected by [28, 29]. This dimorphism may have developed multiple factors secondary to ACTH stimulation to deal with the exposure of each sex to a [202]. The hypothalmus-pituitary-adrenal sys- different steroid spectrum. tem can also be stimulated by cytokines, e.g. Daily fluctuations in the levels of ACTH are interleukin-l, produced by the cells of the not immediately associated with changes in immune system [203]. Rapid increases in steroid enzyme levels; yet, this daily intermittent stimu- secretion in response to ACTH or other factors, lation of the adrenal cortex is essential for the probably as a rule, result from stimulation of maintenance of the normal levels of the steroidogenesis via increased cholesterol mobil- steroidogenic enzyme in these tissues. In hypo- ization and transport into mitochondria [2]. physectomized animals, the activities of many The activation of the hypothalarnus-pituitary- steroidogenic enzymes decrease greatly [17, 129, adrenal axis is vital to cope with stress and 190, 191], in addition to many other cellular disease [201]. Under continued stress, the changes [e.g. 208]. If the duration of trophic constant simulation of the adrenal cortex can hormone deprivation is not prolonged, these enhance its capacity to secrete glucocorticoids changes can be reversed by the administration by increasing the levels of key steroidogenic of trophic hormones [190, 191]. Steroidogenic enzymes 791

The secretion of aldosterone from zona be effects directly on the testicular function glomerulosa is regulated mainly by the renin- [214]. In vivo immune activation of macrophages angiotensin system and blood sodium and that neighbor Leydig cells in the interstitial tissue potassium levels [6-8, 149-151]. Angiotensin II was found to decrease the level of P450c17 in can rapidly stimulate aldosterone secretion from Leydig cells of mice to 10% of control values glomerulosa cells by increasing cholesterol [215]. In vitro treatment of Leydig cells with mobilization similar to ACTH stimulation of interleukin-1 similarly decreased P450c17 the zona fasciculata [2]. However, the intra- expression [215]. Interferon-), also inhibits Leydig cellular transduction systems involved in this cell steroidogenesis and hCG induced enzyme rapid stimulation are different from that of expression[216]. Thus, cytokines and other ACTH [151, 211]. factors released from immune cells may affect In rats fed a low sodium or high potassium testicular steroidogenic output by suppressing diet, plasma aldosterone levels increase to main- the expression of enzymes essential for androgen tain a normal level of plasma electrolytes [150]. synthesis. The maintenance of this diet for several days, enhances the activity, the protein level, and the 4.5. Ovary mRNA level of P450scc and P450c18 while In contrast to the adrenal cortex and testis, the same parameters for P450cl 1 remain with- the blood levels of ovarian steroid hormones out a significant change [149, 150]. These results change greatly during the female reproductive establish that constant stimulation of zona cycle, which can last many days. The estrous glomerulosa activity enhances its steroidogenic cycles of rodents and ruminants, and the capacity by increasing specific enzyme levels in menstrual cycles of primates are characterized zona glomerulosa and not in zona fasciculata by stages of ovarian follicle development, ovula- [149, 150]. tion, corpus luteum formation and subsequent luteolysis and regression in the absence of preg- 4.4. Testis nancy. The lengths of these stages vary greatly Similar to adrenocortical steroids, the plasma among species, yet, the pattern of steroid hor- levels of the testicular steroids fluctuate in re- mone levels during the reproductive cycle of sponse to diurnal variations in trophic hormone most mammalian species shows certain com- gonadotropin secretion [197]. Studies on hypo- mon characteristics: (1) a sharp rise in estradiol physectomized animals indicate that this daily levels prior to the ovulatory surge of LH. (2) An intermittent stimulation by gonadotropins is increase in progesterone levels after ovulation necessary for the expression of steroidogenic that is sustained during the luteal phase or enzymes [190, 191,212]. Yet, the degree of this ensuing pregnancy (human [217], monkeys dependence varies for different enzymes [212]. [218-220], cow[221], rat[222]). The studies, In different strains of inbred mice the steroido- summarized below, indicate that these changes genie capacity of the Leydig cells is highly result from major changes in the levels of the correlated with the levels of P450scc [209]. steroidogenic enzymes during the development In hypogonadal mice with a defective gonado- of the ovarian follicle. tropin releasing hormone gene, and consequently The rise in estradiol levels prior to ovulation undetectable levels of LH and FSH, the activities is dependent on LH/FSH stimulation of the of most steroidogenic enzymes, except 3fl-HSD, final stages of preovulatory follicle development are very low or undetectable in the testis [213]. [134]. As noted above, estradiol biosynthesis LH treatment of these mice increases the by P450arom in granulosa cells is dependent testieular enzyme activities within 10 days. on androgen precursor from the theca interna These results provide additional evidence for the (Table 3). Thus, to increase estradiol biosynthe- dependence of steroidogenic enzyme expression sis, the following changes would be expected to on gonadotropin stimulation, and reveal that occur in the follicle: a rise in theca cell enzymes exposure to gonadotropins during ontogeny is responsible for androgen biosynthesis, e.g. not essential for the development of this capacity P450scc and P450c17, and a rise in granulosa [213]. cell P450arom. Studies on rats indeed showed In contrast to adrenal cortex, stress and patho- that gonadotropin stimulation greatly enhances logical conditions often disrupt the function of P450scc levels in thecal and interstitial tissue the hypothalamic-pituitary-testicular axis by within 24 h, while granulosa cells remain with- suppressing LH [214]. However, there may also out detectable P450scc[223]. The level of

SBMB 43/&--E 792 ISRAEL HANUKOGLU

P450c17 is correlated with the developmental which have been shown to also enhance their stage/size of preovulatory follicles [135]. In vitro expression in granulosa cells in culture, ([154]; studies with human and rat granulosa cells human [137, 233, 234], bovine [235], porcine showed that gonadotropin stimulation greatly [236, 237], rat [229,238], chicken [238a]. increases P450arom mRNA, protein level and However, after this initial turning-on, the activity [109, 224]. These studies establish that expression of these genes can continue in luteal the preovulatory rise of estradiol is a result of cells in the absence of gonadotropin stimulation enhancement of expression of specific steroido- [68, 154, 229]. genic enzymes in a defined population of cells in One interesting aspect of steroidogenic the preovulatory follicle. enzyme expression in the developing follicle The increase in blood progesterone levels is the vectorial change that is initiated by after ovulation, results from two concomitant the gonadotropins. During the final stages of processes: growth of the corpus luteum, and an maturation of the rat preovulatory follicle, increase in steroidogenic enzymes. Progesterone P450scc is expressed first at the periphery of biosynthesis is dependent on two enzymes the follicle in theca interna cells and then in P450scc, and 3fl-HSD (Fig. 1). The expression granulosa cells and finally, just before ovula- of these enzymes is greatly enhanced in tion, in cumulus cells [223, 241]. The sequential luteal cells (cow [121,225-227], pig [122], rat [17, expression of P450scc in the different follicular 135,228, 229]). Expression of P450scc in rat cells could result from a gradient of gonado- granulosa cells is initiated with a delay after tropin concentration or responsiveness in the expression in theca cells, and takes place close follicle. In the rat preovulatory follicle, the to the time of LH surge [223,229]. After ovula- concentration of LH receptors shows a steep tion, the theca and granulosa cells become the gradient decreasing from the periphery of the progenitors of small and large luteal cells of the follicle to its center [242, 243]. Immunohisto- corpus luteum, while this cell lineage continues chemical and in situ hybridization studies re- to be reflected in the functioning of the two vealed that LH receptors are expressed only in types of luteal cells [230-232, 291]. The ovula- granulosa cells of preovulatory follicles, but not tory surge of LH is associated with the disap- in small follicles [291,292]. Thus, the expression pearance of P450c17 expression in bovine [225] of P450scc in granulosa cells is closely associ- and rat [135], but not in human corpora lutea. ated with the induction of LH receptors in these The factor(s) that mediate this suppression may cells. be different from LH, because in cultured The steroidogenic response of follicular cells luteinizing rat follicles the activity of P450c17 is to gonadotropins may be modulated by other maintained with or without LH [135, 154]. The endocrine and paracrine factors, e.g. ovarian continued expression of P450arom for estrogen steroids, inhibin, activin, and various peptide synthesis in the corpus luteum varies among factors [138, 244, 245]. Among these, estradiol species [109, 138, 230]. and -like growth factors (IGF) have been In the corpus luteum, the total amount of shown to have a strong synergistic effect with P450scc can increase about 100-fold over the gonadotropins on the induction of P450scc in levels found in the ovary (Table 5). This is granulosa cells from some species [67, 236, 246]. accompanied by increases in the levels of the In human granulosa cells, IGF-II and P450scc electron carriers adrenodoxin reductase and mRNAs are coordinately regulated [233]. IGFs adrenodoxin [121,122]. In contrast, the level of probably function mainly as paracrine factors, the microsomal P450 reductase shows no mediating some actions of trophic hormones, significant change [225]. The increases in the as they are secreted by ovarian cells, and can act levels of P450scc and 3fl-HSD are correlated locally on granulosa cells that have receptors for with increases in their mRNAs, indicating IGF [245]. Vasoactive intestinal peptide was enhanced transcription of the respective shown to induce P450scc in rat granulosa cells genes [226, 227]. The correlation of the levels though to a very minor degree as compared to of the three mitochondrial P450scc system FSH [247]. proteins[121] indicate that the genes of Ovarian cells possess a system of cholesterol these enzymes may share certain common mobilization similar to adrenocortical cells [237, regulatory sequences which remain to be ident- 248, 249]. Thus, gonadotropin stimulation of ified. The change in the levels of these enzymes cholesterol mobilization is required in addition to is probably directly initiated by gonadotropins effects on enzyme expression. Steroidogenic enzymes 793

4.6. Mechanisms of regulation of steroidogenic cDNA probes, respectively), enzyme levels enzyme levels increased only between 6-12h after ACTH stimulation of adrenocortical cells, a time when The studies summarized above indicate that mRNA levels had already reached their peak the level of expression of each steroidogenic values [38]. Thus, the increase in enzyme level enzyme varies in three characteristics: lags behind the mRNA level by 6-12h[38]. (a) Tissue- and cell-specific expression, deter- When the mRNA levels start to drop, the levels mined by processes that switch genes on of the enzymes do not show an accompanying or off during tissue and cell differen- decrease, probably reflecting the fact that the tiation, fixing the fate of the regulation of enzymes have much longer half lives than the the gene in successive lineages of the cell. mRNAs (Section 4.1). The time course and (b) Basal expression, determined by intra- magnitude of induction of mRNAs encoding cellular processes in the absence of trophic different enzymes vary significantly, indicating hormonal stimulation. differential regulation of the genes (see Ref. [38]). (c) Hormonal signal regulated expression, The reason for the relative slowness of the determined by hormonal stimuli that induction of the steroidogenic enzyme mRNAs affect the expression of the gene positively is currently not known. The mechanism of this or negatively. induction is probably different from that of other genes that can be induced rapidly, within Each of these three types of expression < 1 h [252]. In some steroidogenic cells, the probably represent the functioning of specific induction of P450 mRNA synthesis can be inhib- gene regulatory elements. ited by cycloheximide, implicating a requirement As noted above, in adult steroidogenic tissues, for the synthesis of a specific protein that may the levels of most of the cell- and tissue-specific be necessary for the induction [250, 253, 254]. steroidogenic enzymes depend mainly on trophic This effect has not been consistently observed in hormonal stimulation, independent of whether all cases of steroidogenic P450 induction [255, the cell's repertoire of enzymes remains constant 256]. Trophic hormones ACTH, angiotensin II, (as in the adrenal cortex and testis) or changes and hCG induce rapidly (in < 1 h) the tran- as a result of tissue growth (as in the ovarian scription of genes encoding c-fos and jun-B follicle) (see Section 4.1). In contrast, during in steroidogenic cells[257]. These 'primary ontogenic development, the same trophic response' gene products function as transcription hormones may not be necessary for the initial factors [258], and may be involved in the regu- expression of enzymes [213, 250]. lation of expression of steroidogenic enzymes. In adult tissues, trophic hormones may affect Enzyme levels may also be affected by a steroidogenic enzyme synthesis via effects on change in the stability or translation rate of the transcription, stability or translation of the mRNAs that encode them. In bovine adreno- mRNAs that encode the enzymes. The major cortical cells in culture, ACTH was observed to physiological variations in enzyme levels (as in increase the stability of P450scc mRNA 5-fold ovarian follicular and adrenal glomerulosa cells, but had no effect on P450c11, P450c17, and see above) are associated with parallel changes P450c21 mRNAs [192]. In in vivo studies with in mRNA levels. The dependence of enzyme guinea pigs, chronic ACTH treatment increases synthesis on transcriptional induction is most the activities of steroidogenic enzymes, without conspicuous in ovarian granulosa cells during a significant effect on the levels of the mRNAs follicle growth (see Section 4.5). In vitro studies encoding the enzymes [133]. This suggests that provide further evidence that trophic hormones under this chronic treatment, ACTH may have can enhance the transcription of genes encoding a post-transcriptional effect on the mRNAs steroidogenic enzymes: after trophic hormonal encoding steroidogenic enzymes [133]. stimulation of steroidogenic cells in culture, mRNA levels for steroidogenic enzymes gener- 4.Z Intracellular regulators of steroidogenic ally increase within a few hours, and reach peak enzyme expression values at 6-12 h (adrenal cortex [38, 39, 90, Trophic hormones regulate the function of 99, 132, 147, 234, 248], ovary [68, 153, 224, 229, steroidogenic cells by binding to specific 235-239] testis [40, 119, 251]). receptors on the cell surface and activating intra- In a study that measured both P450scc system cellular signal transduction systems [259-263]. enzyme and mRNA levels (using antibody and Trophic hormone receptors that have been 794 ISRAEL HANUKOGLU characterized to-date belong to the superfamily in cAMP activated protein kinase A alters of G-protein linked receptors with seven hydro- enzyme inducibility in a mouse adrenocortical phobic segments embedded in the cell membrane cell line [282]. [264, 265]. The interaction of hormone bound The criterion of similar duration of action with specific G-proteins [266] activates (see above) was examined only in a recent study some membrane bound enzymes, e.g. adenylate that compared the effects of ACTH and cAMP cyclase and phospholipases, causing a transient [38]. Normally, ACTH is secreted in pulses with increase in intracellular levels of second messen- a circadian pattern and the blood level of ACTH gers such as cAMP, cGMP, triphos- shows a peak that lasts only a few hours [198]. phate, Ca 2+, and diacylglycerol. These small The intracellular levels of cAMP peak even regulatory molecules transmit the hormonal sig- more rapidly, within 5-10 min after hormonal nal inside the cell by activating protein kinases, stimulation, and then gradually decrease [38, phospholipases, or by directly interacting with 283]. In an experiment designed to mimic this other proteins. For example, cAMP activates physiological pattern, adrenocortical cells were protein kinase A, and Ca ~+ and diacylgycerol exposed to ACTH, cAMP analogs, or forskolin, activate protein kinase C [259-262]. An addi- for only short periods, and the cells were tional signal transduction system involves tyro- harvested after 12-36h to assay enzyme and sine kinases, which are generally activated by mRNA levels. Whereas, a short exposure (30- growth factors, but also may be involved in the 60 min) of cells to ACTH induced the enzymes actions of some hormones [263]. Some of these and their mRNAs, cAMP induction required different signaling systems may be activated in treatment of cells continuously for nearly 24 h concert to modulate the cellular response by a [38]. These findings question the physiological complex crosstalk network [259-262]. The ulti- relevance of experiments using prolonged (24 h) mate cellular response to hormones is generally stimulation with cAMP analogs, and suggest effected by protein kinases, which phosphorylate that trophic hormone action is not mediated proteins and affect their function [259-262]. solely through cAMP but uses additional signal In examining possible molecules that may transduction systems. mediate the effects of trophic hormones, the fol- The cAMP pathway of enzyme induction may lowing questions are posed: (1) Does the agent be modified by the activation of other signaling have the same effect as the hormone? (2) Does systems. As noted in Section 4.5, IGF-I has a the trophic hormone increase the levels of the synergistic effect with gonadotropins on the in- agent at doses similar to those required for the duction of P450scc in granulosa cells. Similarly, cellular response? (3) Are the time courses in secondary cultures of adrenocortical cells and durations of action of the agent and the the induction of lift- and 21-hydroxylases by hormone similar? (4) Does inhibiting the action cholera toxin, but not of 17~-hydroxylase, is of the agent inhibit the action of the hormone? dependent on IGF-I [284, 285]. A tyrosine kinase Based on some of these criteria, the following inhibitor, tryphostin, arrests FSH or cAMP agents have been suggested as mediators of induced synthesis of P450scc in granulosa rapid hormonal activation of steroidogenesis cells, suggesting that a tyrosine kinase may be via cholesterol mobilization: cAMP, cGMP, involved in the pathway of signal transduction inositol triphosphate, Ca 2+, diacylglycerol and downstream from cAMP [286]. arachidonic acid [211,267-281]. Besides protein kinase A, both adrenocortical In both adrenocortical and gonadal cells, and gonadal cells possess protein kinase C cAMP has been viewed as the main mediator of [287, 288]. The concurrent activation of protein trophic hormone (ACTH and LH/FSH, respect- kinase A and C pathways, (using agents that ively) induction of steroidogenic enzymes, based increase cAMP, and phorbol ester, TPA, respec- on the following observations: (a) these hor- tively) generally has been observed to suppress mones stimulate adenylate cyclase and increase expression of steroidogenic enzymes in both intracellular cAMP (refs in previous paragraph); adrenocortical cells [61,284, 289] and a granu- (b) treatment of cultured cells for > 24 h with losa cell line where P450scc can be induced by cAMP analogs, or other agents (e.g. forskolin, cAMP analogs [167, 290]. In the granulosa cells, cholera toxin) that increase intracellular cAMP, TPA shows no effect by itself, but severely induces expression of steroidogenic enzyme inhibits induction of steroidogenesis when added mRNAs to the same levels as hormonal stimu- with forskolin [287]. TPA does not inhibit fors- lation (refs in Sections 4.3-4.5); (c) a mutation kolin elevation of cAMP levels, indicating that Steroidogenic enzymes 795

TPA prevents the action, but not the synthesis and R. B. Jaffe). W. B. Saunders, Philadelphia, 3rd Edn of cAMP [287]. The effect of TPA on P450e21 (1991) pp. 156-180. 2. Jefcoate C. R., McNamara B. C., Artemenko I. and expression in adrenocortical cells depends on Yamazaki T.: Regulation of cholesterol movement to the mitotic activity of the cells: in non-dividing mitochondrial cytochrome P4508cc in steroid hormone cultures it is without effect, whereas in cultures synthesis. J. Steroid Bioehem. Molec. Biol. 43 (1992) 751-767. with high mitotic activity it induces P450c21 at 3. Lambeth J. D. and Stevens V. L.: Cytochrome low concentrations (0.3-3 nM TPA), and inhibits P-450sec: Enzymology, and the regulation of intra- expression at a higher concentration [285]. Even mitochondrial delivery to the enzyme. Endocrine Res. 10 (1985) 283-309. extensive exposure to ACTH does not stimulate 4. Orme-Johnson N. R.: Distinctive properties of adrenal adrenocortical cell proliferation in vivo [208]. cortex mitochondria. Biochim. Biophys. Acta 1020 Thus, effects in cultures undergoing mitosis (1990) 213-231. 5. Miller W. L. and Levine L. S.: Molecular and clinical probably do not reflect the normal function of advances in congenital adrenal hyperplasia. J. Pediat. the adult adrenal cortex. The inhibitory effect 111 (1987) 1-17. of protein kinase C activation may be part of a 6. Okamoto M. and Nonaka Y.: Molecular biology of rat steroid 1 l~-hydroxylase [P450(11/~)] arid aldosterone homeostatic mechanism. synthase [P450(llfl, ALDO)]. J. Steroid Biochem. To summarize the current results it can be Molec. Biol. 41 (1992) 3-8. concluded that while cAMP pathway may be 7. Shizuta Y., Kawamoto T., Mitsuuchi Y., Toda K., Miyahara K., Ichikawa Y., Imura H. and Ulick S.: part of the mechanism of steroidogenic enzyme Molecular genetic studies on the biosynthesis of aldo- gene induction by trophic hormones, many parts sterone in humans. J. Steroid Biochem. Molec. Biol. 43 of the puzzle are still missing. There are lines of (1992) 981-987. 8. White P. C., Pascoe L., Curnow K, M., Tannin G. and evidence indicating that the same pathway is not R6sler A.: Molecular biology of 1 lfl-hydroxylase and mediating both rapid cholesterol mobilization 1lfl-hydroxysteroid dehydrogenase enzymes. J. Steroid and slow enzyme induction. The pathways for Biochem. Molec. Biol. 43 (1992) 827-835. 9. Rosier A. and E. Leiberman: Enzymatic defects of these distinct processes may have a common steroidogenesis: 1lfl-hydroxylase deficiency congenital origin but we do not yet know where and how adrenal hyperplasia. In Pediatric and Adolescent they diverge. Elucidation of the mechanism of (Edited by Z. Laron). S. Karger, Basel, Vol. 13 (1984) pp. 47-71. enzyme induction requires an understanding of 10. Pascoc L., Curnow K. M., Slutsker L., Rosier A. how brief pulses of trophic hormones can exert and White P. C.: Mutations in the human CYPi IB2 long lasting effects [e.g. 38]. These actions may (aidosterone synthase) gene causing corticosterone methyloxidase II deficiency. Proc. Natn. Acad. Sci. be mediated by a cascade of short-lived medi- U.S.A. 89 (1992) 4996-5000. ators (e.g. cAMP ---, protein kinase A ---, protein 11. Takemori S. and Kominami S.: Adrenal microsomal phosphorylation --.---, primary response gene cytochrome P-450 dependent reactions in steroido- genesis and biochemical properties of the enzymes induction--, cellular response), or alternatively involved therein. In Frontiers in Biotransformation by as yet unidentified long-lasting mediators (Edited by K. Ruckpaul and H. Rein). Akademie, that after initial hormonal stimulation maintain Verlag Berlin. 3 (1991) pp. 153-203. 12. Yanase T., Imai T., Simpson E. R. and Waterman a metabolic memory in the cell. Future research M. R.: Molecular basis of 17,,-hydroxylase/17,20-1yase coming from two directions, investigating events deficiency. J. Steroid Biochem. Molec. Biol. 43 (1992) from the cell membrane towards the 973-979. 13. Miller W. L., Gitelman S. E., Bristow J. and Morel Y.: on the one hand, and from gene regulatory Analysis of the duplicated human C4/P450c21/X gene elements towards the cytoplasmic regulatory cluster. J. Steroid Biochem. Molec. Biol. 43 (1992) factors on the other, gives hope to provide the 961-971. 14. White P. C. and New M. 1.: Genetic basis of Endocrine answers. disease 2: congenital adrenal hyperplasia due to 21- hydroxylase deficiency. J. Clin. Endocr. Metab. 74 Acknowledgements--I am most grateful to Drs A. (1992) 6-11. Hanukoglu, A. M. Kaye, J. Orly, M. Raikhinstein and 15. Santen R. J.: : future perspectives. Steroids A. Rosier and Ms M. Zohar for their valuable comments on 50 (1987). the manuscript. Our research reviewed here was supported 16. Simpson E. R., Kilgure M. W., Mahendroo M. S., by grants from the U.S. National Institutes of Health, The Means G. D., Corbin C. J. and Mendelson C. R.: Leo and Julia Forchheimer Center for Molecular Genetics, Regulation of human aromatase The Joseph and Ceil Mazer Center for Structural Biology, gene expression. 3'. Steroid Biochem. Molec. Biol. 43 and The Dr Josef Cohn Center for Biomembrane Research (1992) 923-930. at the Weizmann Institute of Science. The author is the 17. Labrie F., Simard J., Luu-The V., B~langer A. and incumbent of the Delta Research Career Development Chair. Pelletier G.: Structure, function and tissne-specific gene expression of 3fl-hydroxysteroid dehydrogenase/5-ene- 4-ene enzymes in classical and peripheral REFERENCES intracrine steroidogenic tissues. J. Steroid Biochem. Molec. Biol. 43 (1992) 805-826. 1. O'Malley B. W. and Strott, C. A.: Steroid hormones. 18. Inano H., Ishii-Ohba H., Sugimoto Y., Ohta Y., In Reproductive Endocrinology (Edited by S. S. C. Yen Morikawa T., Yoshida M. and Tamaoki B.: 796 ISRAEL HANUKOGLU

Purification and properties of enzymes related to ster- 34. Burchell B. and Coughtrie W. H.: UDP-glucuronosyl- oid hormone synthesis. Ann. N.Y. Acad. Sci. 595 . Pharmac. Ther. 43 (1989) 261-289. (1990) 17-25. 35. Mackenzie P. I., Rodbourne L. and Stranks S.: Steroid 19. Luu-The V., Labile C., Zhao H.-F., Couet J., UDP giucuronosyltransferases. J. Steroid Biochem. Lachance Y., Simard J., Cote J., Leblanc G., Lagace Melee. Biol. 43 (1992) 1099-1105. L., Berube D., Gagne R. and Labile F.: Purification, 36. Henry F. J. and Bassett J. R.: Corticosterone storage cloning, complementary DNA structure, and predicted within the adrenal cortex: Evidence for a sulphate sequence of human estradiol 17p-dehydro- conjugate. J. Endocr. 104 (1985) 381-386. genase. Ann. N.Y. Acad. Sci. 595 (1990) 40-52. 37. Bclanger B., Couture J., Caron S., Bodou, P., Fiet J. 20. Martel C., Rheaume E., Takahashi M., Trudel C., and Belanger A.: Production and secretion of C19 Couet J., Luu-The V., Simard J. and Labile F.: steroids by rat and guinea pig adrenals. Steroids 55 Distribution of 17//-hydroxysteroid dehydrogenase (1990) 360-365. gene expression and activity in rat and human tissues. 38. Hanukoglu I., Feuchtwanger R. and Hanukoglu A.: J. Steroid Biochem. Molec. Biol. 41 (1992) 597- Mechanism of ACTH and cAMP induction of mito- 603. chondrial cytochrome P450 system enzymes in adrenal 21. R6sler A.: Steroid 17fl-hydroxysteroid dehydrogenase cortex cells. J. Biol. Chem. 265 (1990) 20602-20608. deficiency in man: an inherited form of male pseudo- 39. Hornsby P. J., Cheng C. Y., Lala D. S., Maghsoudlou hermaphroditism. J. Steroid Biochem. Molec. Biol. 43 S. S., Raju S. G. and Yang L.: Changes in gene (1992) 989-1002. expression during senescence of adrenocortical cells in 22. Archakov A. I. and Bachmanova G. I.: Cytochrome culture. J. Steroid Biochem. Molec. Biol. 43 (1992) P-450 and Active Oxygen. Taylor & Francis, U.K. 951-960. (1990). 40. Payne A. H., Youngblood G. L., Sha L., Burgos- 23. Guengerich F. P. (Ed.): Mammalian Cytochromes Trinidad M. and Hammond S. H.: Hormonal regula- P-450. CRC Press, Boca Raton, FL. Vols 1 & 2 (1987). tion of steroidogenic enzyme gene expression in Leydig 24. Hanioka N., Gonzalez F. J., Lindberg N. A., Liu G. cells. J. Steroid Biochem. Melee. Biol. 43 (1992) and Korzekwa K. R.: Chimeric eDNA expression and 895-906. site directed mutagenesis studies of cytochrome P450s 41. Schimmer B. P. and Parker K. L.: Promoter elements CYP2A1 and CYP2A2. J. Steroid Biochem. Melee. of the mouse 21-hydroxylase (Cyp-21) gene involved in Biol. 43 (1992) 1037-1043. cell-selective and cAMP-dependent gene expression. 25. Nebert D., Nelson D. R., Coon M. J., Estabrook J. Steroid Biochem. Melee. Biol. 43 (1992) 937-950. R. W., Feyereisen R., Fujii-Kuriyama I., Gonzalez 42. Waterman M. R., Kagawa N., Zanger U. M., Momoi F. J., Guengerich F., Gunsalus I. C., Johnson E. F., K., Land J. and Simpson E. R.: Comparison of cAMP- Loper J. C., Sate R., Waterman M. R. and Waxman responsive DNA sequences and their binding proteins D. J.: The P450 superfamily: update on new sequences associated with expression of the bovine CYP17 and gene mapping, and recommended nomenclature. CYPllA and human CYP21B genes. J. Steroid DNA Cell Biol. 10 (1991) 1-14. Biochem. Molec. Biol. 43 (1992) 931-935. 26. Negishi M., Iwasaki M., Juvonen R. O. and Aida K.: 43. Hall P. F. and Almahbobi G.: The role of cytoskeleton Alteration of the substrate specificity of mouse 2A in the regulation of steroidogenesis. J. Steroid Biochem. P450s by the identity of residue-209: steroid-binding Melee. Biol. 43 (1992) 769-777. site and orientation. J. Steroid Biochem. Melee. Biol. 44. Armbrecht H. J., Okuda K., Wongsurawat N., 43 (1992) 1031-1036. Nemani R. K., Chen M. L. and Boltz M. A.: Charac- 27. Schenkman J. B.: Steroid metabolism by constitutive terization and regulation of the vitamin D hydroxyl- cytochromes P450. J. Steroid Biochem. Melee. Biol. 43 ases. J. Steroid Biochem. Molec. Biol. 43 (1992) (1992) 1023-1030. 1073-1081. 28. Waxman D. J.: Regulation of liver-specific steroid 45. Shemesh M.: Production and regulation of progester- metabolizing cytochromes P450: cholesterol 7~- one in bovine corpus luteum and placenta in mid and hydroxylase, 6fl-hydroxylase, and growth late gestation: a personal review. Reprod. Fert. Devel. hormone-responsive steroid hormone hydroxylases. 2 (1990) 129-135. J. Steroid Biochem. Melee. Biol. 43 (1992) 1055-1072. 46. Simpson E. R. and MacDonald P. C.: Endocrine 29. Westin S., Toilet P., Str6m A., Mode A. and Gustafsson physiology of the placenta. A. Rev. Physiol. 43 (1981) J. A.: The role and mechanism of in 163-188. the regulation of sexually dimorphic P450 enzymes in 47. Hanukoglu I.: Reductases of the bacterial, mito- rat liver. J. Steroid Biochem. Melee. Biol. 43 (1992) chondrial and microsomal cytochrome P450 systems: 1045-1053. structural comparisons. In Cytochrome P-450: Bio- 30. Cheng K.-C.: Molecular cloning of rat liver 3a-hydroxy- chemistry and Biophysics (Edited by A. I. Archakov steroid dehydrogenase and identification of structurally and G. I. Bachmanova). INCO-TNC, Moscow (1992) related proteins from rat lung and kidney. J. Steroid pp. 339-344. Biochem. Molec. Biol. 43 (1992) 1083-1088. 48. Hanukoglu I., Gutfinger T., Haniu M. and Shively 31. Jenkins E. P., Andersson S., Imperato-McGinley J., J. E.: Isolation of a eDNA for adrenodoxin reductase Wilson J. D. and Russell D. W.: Genetic and pharmaco- (ferredoxin--NADP + reductase): implications for logical evidence for more than one human steroid mitochondrial cytochrome P-450 systems. Eur. J. 5a-reductase. J. Clin. Invest. 89 (1992) 293-300. Biochem. 169 (1987) 449-455. 32. Nakajin S., Fujii S., Ohno S. and Shinoda M.: 49. Lin D., Shi Y. and Miller W. L.: Cloning and sequence 3a-Hydroxysteroid dehydrogenase activity catalyzed of the human adrenodoxin reductase gene. Prec. Nam. by purified pig adrenal 20a-hydroxysteroid dehydro- Acad. Sci. U.S.A. 87 (1990) 8516-8520. genase. J. Steroid Biochem. Melee. Biol. 41 (1992) 50. Solish S. B., Picado-Leonard J., Morel Y., Kuhn 179-184. R. W., Mohandas T. K., Hanukogiu I. and Miller 33. Demyan W. F., Song C. S., Kim D. S., Her S., Gallwitz W. L.: Human adrenodoxin reductase--Two mRNAs W., Rao T. R., Slomczynska M., Chatterjee B. and encoded by a single gene on chromosome 17cen-q25 Roy A. K.: Estrogen of the rat liver: are expressed in steroidogenic tissues. Prec. Natn. Complementary DNA cloning and age- and sex-specific Acad. Sci. U.S.A. 85 (1988) 7107-7108. regulation of messenger RNA. Molec. Endocr. 6 (1992) 51. Sparkes R. S., Klisak I. and Miller M. L.: Regional 589-597. mapping of genes encoding human steroidogenic Steroidogenic enzymes 797

enzymes: P450scc 15q23-q24 adrenodoxin to esterol side-chain cleavage eDNA isolated from porcine 11q22; Adrenodoxin reductase to 17q24-q25; and testis. Nucleic Acids ICes. 17 (1989) 1773. P450c17 to 10q24-q25. DNA Cell Biol. I0 (1991) 67. McMasters K. M., Dickson L. A., Shamy R. V., 359-365. Robischon K., Macdonald G. J. and Moyle W. R.: Rat 52. Hanukoglu I. and Gutfinger T.: eDNA sequence of cholesterol side-chain cleavage enzyme (P-450scc): use adrenodoxin reductase: Identification of NADP binding of a cDNA probe to study the hormonal regulation sites in oxidoreduetases. Eur. J. Biochem. 180 (1989) of P-450scc mRNA levels in ovarian granulosa cells. 479-484. Gene 57 (1987) 1-9. 53. Sagara Y., Takata Y., Miyata T., Hara T. and 68. Oonk R. B., Krasnow J. S., Beattie W. G. and Horiuchi T.: Cloning and sequence analysis of adreno- Richards J. S.: Cyclic AMP-dependent and inde- doxin reductase eDNA from bovine adrenal cortex. pendent regulation of cholesterol side chain cleavage J. Biochem. 102 (1987) 1333-1336. cytochrome P-450 in rat ovarian granulosa cells and 54. Chang C. Y., Wu D. A., Mohandas T. K. and Chung corpora lutea, eDNA and deduced amino acid sequence B. C.: Structure, sequence, chromosomal location, of rat P-450see. J. Biol. Chem. 264 (1989) 21934- and evolution of the human ferredoxin gene family. 21942. DNA Cell Biol. 9 (1990) 205-212. 69. Mornet E., Dupont J., Vitek A. and White P. C.: 55. Picado-Leonard J., Voutilainen R., Kao L., Chung B., Characterization of two genes encoding human steroid Strauss III J. F. and Miller W. L.: Human adreno- 1l~-hydroxylase (P-45011~0). J. Biol. Chem. 264 (1986) doxin: Cloning of three cDNAs and cycloheximide 20961-20967. enhancement in JEG-3 cells. J. Biol. Chem. 263 (1988) 70. Kawamoto T., Mitsuuchi Y., Toda K., Miyahara K., 3240-3244 (correction on p. 11016). Yokoyama Y., Nakao K., Hosoda K., Yamamoto Y., 56. Hiwatashi A., Sakihama N., Shin M. and Ichikawa Y.: Imura H. and Shizuta Y.: Cloning of eDNA and Heterogeneity of adrenocortical ferredoxin. FEBS Left. genomic DNA for human cytochrome P-45011/L 209 (1986) 311-315 FEBS Lett. 269 (1990) 345-349. 57. Okamura T., John M. E., Zuber M. X., Simpson E. R. 71. Morohashi K., Yoshioka H., Gotoh O., Okada T., and Waterman M. R.: Molecular cloning and amino Yamamoto K., Miyata T., Sogawa K., Fujii-Kuriyama acid sequence of the precursor form of bovine adreno- Y. and Omura T.: Molecular cloning and nucleotide doxin: Evidence for a previously unidentified COOH- sequence of DNA of mitochondrial cytochrome P- terminal peptide. Proc. Natn. Acad. Sci. U.S.A. 82 450(11/1) of bovine adrenal cortex. J. Biochem. 102 (1985) 5705-5709. (1987) 559-568. 58. Okumura T., Kagimoto M., Simpson E. R. and 72. Hashimoto T., Morohashi T. and Omura T.: Cloning Waterman M. R.: Multiple species of bovine adreno- and characterization of bovine cytochrome P-450(1 l/I) doxin mRNA. Occurrence of two different mito- genes. J. Biochem. 105 (1989) 676-679. chondrial precursor sequences associated with the same 73. Kirita S., Morohashi K., Hashimoto T., Yoshioka H., mature sequence. J. Biol. Chem. 262 (1987) 10335- Fujii-Kuriyama Y. and Omura T.: Expression of two 10338. kinds of cytochrome P-450(I I/]) mRNA in bovine 59. Sagara Y., Sawae H., Kimura A., Sagara-Nakano Y., adrenal cortex. J. Biochem. 104 (1988) 683-686. Morohashi K., Miyoshi K. and Horiuchi T.: Struc- 74. Kirita S., Hashimoto T., Kitajima M., Honda S., tural organization of the bovine adrenodoxin gene. Morohashi T. and Omura T,: Structural analysis of J. Biochem. 107 (1990) 77-83. multiple bovine P-450(I 1/]) genes and their promoter 60. Chung B., Matteson K. J., Voutilainen R., Mohandas activities. J. Biochem. 108 (1990) 1030-1041. T. K. and Miller W. L.: Human cholesterol side-chain 75. Nonaka Y., Matsukawa N., Morohashi K., Omura T., cleavage enzyme, P450scc: eDNA cloning, assignment Ogihara T., Teraoka H. and Okamoto M.: Molecular of the gene to chromosome 15, and expression in the cloning and sequence analysis of eDNA encoding rat placenta. Proc. Natn. Acad. Sci. U.S.A. 83 (1986) adrenal cytochrome P-450u~. FEBS Left. 255 (1989) 8962-8966. 21-26. 61. Moore C. C. D., Brentano S. T. and Miller W. L.: 76. Domalik L. J., Chaplin D. D., Kirkman M. S., Wu Human P450~6c gene transcription is induced by cyclic R. C., Liu W., Howard T. A., Seldin M. F. and Parker AMP and repressed by 12-O-tetradecanoylphorbol-13- K. L.: Different isozymes of mouse 1 l~-hydroxylase acetate and A23187 through independent cis elements. produce mineralocorticoids and glucocorticoids. Molec. Molec. Cell. Biol. 10 (1990) 6013-6023. Endocr. $ (1991) 1853-1862. 62. Morohashi K., Sogawa K., Omura T. and Fujii- 77. Kawamoto T., Mitsuuchi Y., Ohnishi T., Ichikawa Y., Kuriyama Y.: G-erie structure of human cytochrome Yokoyama Y., Sumimoto H., Toda K., Miyahara K., P-450(SCC), cholesterol desmolase. J. Biochem. 101 Kuribayashi I., Nakao K., Hosoda K., Yamamoto Y., (1987) 879--887. Imura H. and Shizuta Y.: Cloning and expression of 63. John M. E., John M. C., Ashley P., MacDonald R. J., a eDNA for human cytochrome P-450aldo as related Simpson E. R. and Waterman M. R.: Identification to primary aldosteronism. Biochem. Biophys. Res. and characterization of eDNA clones specific for Commun. 173 (1990) 309-316. cholesterol side-chain cleavage cytochrome P-450. 78. Matsukawa N., Nonaka Y., Ying Z., Higaki J., Proc. Natn. Aead. Sci. U.S.A. 81 (1984) 5628-5632. Ogihara T. and Okamoto M.: Molecular cloning and 64. Morohashi K., Fujii-Kuriyama Y., Okada Y., Sogawa expression of cDNAs encoding rat : K., Hirose T., Inayama S. and Omura T.: Molecular variants of eytochrome P-45011~. Biochem. Biophys. cloning and nucieotide sequence of eDNA for mRNA Res. Commun. 169 (1990) 242-252. of mitochondrial cytochrome P-450(SCC) of bovine 79. Simmons D. L., Lalley P. A. and Kasper C. B.: adrenal cortex. Proc. Nam. Aead. Sci. U.S.A. gl (1984) Chromosomal assignments of genes coding for com- 4647-4651. ponents of the mixed function oxidase system in mice. 65. Ahlgren R., Simpson E. R., Waterman M. R. and J. Biol. Chem. 260 (1985) 515-521. Lurid J.: Characterization of the promoter regulatory 80. Haniu M., McManus M. E., Birkett D. J., Lee T. D. region of the bovine CYPI 1A (P-450scc) gene. Basal and Shively J. E.: Structural and functional analysis and cAMP-dependent expression. J. Biol. Chem. 265 of NADPH--cytochrome P-450 reductase from human (1990) 3313-3319. liver: complete sequence of human enzyme and 66. Mulheron G. W., Stone R. T., Miller W. L. and Wise NADPH binding sites. Biochemistry 28 (1989) 8639- T.: Nucleotide sequence of cytochrome P-450 chol- 8645. 798 ISRAEL HANUKOGLU

81. Haniu M., Iyanagi T., Miller P., Lee T. D. and Shively gene. Proc. Natn. Acad. Sci. U.S.A. 83 (1986) 2841 J. E.: Complete amino acid sequence of NADPH--cyto- 2845. chrome P-450 reductase in porcine hepatic microsomes. 96. White P. C., New M. I. and Dupont B.: Structure Biochemistry 25 (1986) 7906-7911. of human steroid 21-hydroxylase genes. Proc. Natn. 82. Katagiri M., Murakami H., Yabusaki Y., Sugiyama T., Acad. Sci. U.S.A. 83 (1986) 5111-5115. Okamoto M., Yamano T. and Ohkawa H.: Molecular 97. Chung B., Matteson K. J. and Miller W. L.: cloning and sequence analysis of full-length eDNA for Cloning and characterization of the bovine gene for rabbit liver NADPH-cytochrome P-450 reductase steroid 21-hydroxylase (P-450c21). DNA 4 (1985) mRNA. J. Biochem. 100 (1986) 945-954. 211-219. 83. Gonzalez F. J. and Kasper C. B.: Cloning of DNA 98. Chung B., Matteson K. J. and Miller W. L.: Structure complementary to rat liver NADPH-cytochrome c (P- of a bovine gene for P-450e21 (steroid 2 i-hydroxylase) 450) oxidoreductase and cytochrome P-450b mRNAs. defines a novel cytochrome P-450 gene family. Proc. Evidence that phenobarbital augments transcription of Natn. Acad. Sci. U.S.A. 83 (1986) 4243-4247. specific genes. J. Biol. Chem. 257 (1982) 5962-5968. 99. John M. E., Okamura T., Dee A., Adler B., John 84. Porter T. D. and Kasper C. B.: Coding nucleotide M. C., White P. C., Simpson E. R. and Waterman sequence of rat NADPH-cytochrome P-450 oxido- M. R.: Bovine steroid 21-hydroxylase: Regulation of reductase eDNA and identification of flavin-binding biosynthesis. Biochemistry 25 (1986) 2846-2853. domains. Proc. Natn. Acad. Sci. U.S.A. 82 (1985) I00. Skow L. C., Womack J. E., Petresh J. M. and Miller 973-977. W. L.: Synteny mapping of the genes for 21 steroid 85. Porter T. D., Beck T. W. and Kasper C. B.: hydroxylase, alpha A crystallin, and class I bovine NADPH-cytochrome P-450 oxidoreductase gene leukocyte antigen in cattle. DNA 7 (1988) 143-149. organization correlates with structural domains of the 101. Yoshioka H., Morohashi K., Sogawa K., Yamane M., protein. Biochemistry 29 (1990) 9814-9818. Kominami S., Takemori S., Okada Y., Omura T. and 86. Bradshaw K. D., Waterman M. R., Couch R. T., Fujii-Kuriyama Y.: Structural analysis of cloned eDNA Simpson E. R. and Zuber M. X.: Characterization for mRNA of microsomal cytochrome P-450 (C21) of complementary deoxyribonucleic acid for human which catalyzes steroid 21-hydroxylation in bovine adrenocortical 17~-hydroxylase: A probe for analysis adrenal cortex. J. Biol. Chem. 261 (1986) 4106-4109. of 17~-hydroxylase deficiency. Molec. Endocr. 1 (1987) 102. White P. C., Chaplin D. D., Weis J. H., Dupont B., 348-354. New M. I. and Seidman J. G.: Two steroid 21- 87. Chung B., Picado-Leonard J., Haniu M., Bienkowski hydroxylase genes are located in the murine S region. M., Hall P. F., Shively J. E. and Miller W. L.: Nature 312 (1984) 465-467. Cytochrome P450c17 (steroid 17~-hydroxylase/17,20 103. Chaplin D. D., Galbraith L. J., Seidman J. G., White ): Cloning of human adrenal and testis cDNAs P. C. and Parker K. L.: Nucleotide sequence analysis indicates the same gene is expressed in both tissues. of murine 21-hydroxylase genes: Mutations affecting Proc. Natn. Acad. Sci. U.S.A. 84 (1987) 407-411. gene expression. Proc. Natn Acad. Sci. U.S.A. 83 88. Picado-Leonard J. and Miller W. L.: Cloning and (1986) 9601-9605. sequence of the human gene for P450c17 (steroid 104. Chen S., Besman M. J., Sparkes R. S., Zollman S., 17~-hydroxylase/17,20 lyase): Similarity with the gene Klisak I., Mohandas T., Hall P. F. and Shively J. E.: for P450c21. DNA 6 (1987) 439-448. Human aromatase: cDNA cloning, southern blot 89. Bhasker C. R., Alder B. S., Dee A., John M. E., analysis and assignment of the gene to chromosome Kagimoto M., Zuber M. X., Ahlgren R., Wang X., 15. DNA 7 (1988) 27 38. Simpson E. R. and Waterman M. R.: Structural 105. Harada N.: Cloning of a complete eDNA encoding characterization of the bovine CYP17 (17cc-hydroxyl- human aromatase: Immunochemical identification and ase) gene. Archs Biochem. Biophys. 271 (1989) 479-487. sequence analysis. Biochem. Biophys. Res. Commun. 90. Zuber M. X., John M. E., Okamura T., Simpson E. R. 156 (1988) 725-732. and Waterman M. R.: Bovine adrenocortical cyto- 106. Simpson E. R., Evans C. T., Corbin C. J., Powell F. E., chrome P-45017a. Regulation of gene expression by Ledesma D. B. and Mendelson C. R.: Sequencing of ACTH and elucidation of primary sequence. J. Biol. eDNA inserts encoding aromatase cytochrome P-450 Chem. 261 (1986) 2475-2482. (P-450AROM). Molec. Cell. Endocr. 52 (1987) 267- 91. Fevold H. R., Lorence M. C., McCarthy J. L., Trant 272. J. M., Kagimoto M., Waterman M. R. and Mason 107. Toda K., Terashima M., Mitsuuchi Y., Yamasaka Y., J. I.: Rat P45017~ from testis: characterization of a full Yokoyama Y., Nojima S., Ushiro H., Maeda T., length eDNA encoding a unique Yamamoto Y., Sagara Y. and Shizuta Y.: Alternative capable of catalyzing both A4- and AS-steroid-17,20 - usage of different poly(A) addition signals for two lyase reactions. Molec. Endocr. 3 (1989) 968-975. major species of mRNA encoding human aromatase 92. Namiki M., Kitamura M., Buczko E. and Dufau P-450. FEBS Lett. 24"/ (1989) 371-376. M. L.: Rat testis P-45017~ eDNA: The deduced amino 108. Toda K., Terashima M., Kawamoto T., Sumimoto H., acid sequence expression and secondary structural Yokoyama Y., Kuribayashi I., Mitsuuchi Y., Maeda configuration. Biochem. Biophys. Res. Commun. 157 T., Yamamoto Y., Sagara Y., Ikeda H. and Shizuta Y.: (1988) 705-712. Structural and functional characterization of human 93. Youngblood G. L., Sartorius C., Taylor B. A. and aromatase P-450 gene. Eur. J. Biochem. 193 (1990) Payne A. H.: Isolation characterization, and chromo- 559-565. somal mapping of mouse P450 17~-hydroxylase/ 109. Hickey G. J., Chen S., Besman M. J., Shively J. E., C17-20 lyase. Genomics 10 (1991) 270-275. Hall P. F., Gaddy-Kurten D. and Richards J. S.: 94. Ono H., Iwasaki M., Sakamoto N. and Mizuno S.: Hormonal regulation, tissue distribution and content of eDNA cloning and sequence analysis of a chicken gene aromatase cytochrome P450 messenger ribonucleic acid expressed during the gonadal development and homo- and enzyme in rat ovarian follicles and corpora lutea: logous to mammalian cytochrome P-450cl 7. Gene 66 Relationship to estradiol biosynthesis. Endocrinology (1988) 77-85. 122 (1988) 1426-1436. 95. Higashi Y., Yoshioka H., Yamane M., Gotoh O. and 110. Terashima M., Toda K., Kawamoto T., Kuribayashi Fujii-Kuriyama Y.: Complete nucleotide sequence I., Ogawa Y., Maeda T. and Shizuta Y.: Isolation of two steroid 21-hydroxylase genes tandemly arranged a full-length cDNA encoding mouse aromatase P450. in human chromosome: a pseudogene and a genuine Archs Biochem. Biophys. 285 (1991) 231-237. Steroidogenic enzymes 799

111. McPhaul M. J., Noble J. F., Simpson E. R., genase/A5-A4 isomerase. Endocrinology 130 (1992) Mendelson C. R. and Wilson J. D.: The expression of 553-555. a functional eDNA encoding the chicken cytochrome 126. Dupont E., Zhao H.-F., Rheaume E., Simard J., P-450arom (aromatase) that catalyzes the formation of Luu-The V., Labrie F. and Pelletier G.: Localization of estrogen from androgen. J. Biol. Chem. 263 (1988) 3fl-hydroxysteroid dehydrogenase/AS-A4-isomerase in 16358-16363. rat gonads and adrenal glands by immunocytochemistry 112. Tanaka M., Telecky T. M., Fukada S., Adachi S., and /n situ hybridization. Endocrinology 127 (1990) Chen S. and Nagarna Y.: Cloning and sequence analysis 1394-1403. of the eDNA encoding P-450 aromatase (P-450arom) 127. Goldring N. B. and Ody J.: Concerted metabolism of from a rainbow trout (Oncorhynchus mykiss) ovary; steroid hormones produced by cocultured ovarian cell relationship between the amount of P450arom mRNA types. J. Biol. Chem. 260 (1985) 913-921. and the production of oestradiol-17/] in the ovary. 128. Sasano H., Mason J. I. and Sasano N.: Immunohisto- J. Molec. Endocr. g (1992) 53-61. chemical analysis of cytochrome P-450 17,,-hydroxylase 113. Hanukoglu I. and Jefcoate C. R.: Mitochondilal cyto- in pig adrenal cortex, testis and ovary. Molec. Cell. chrome P-450scc: Mechanism of electron transport Endocr. 62 (1989) 197-202. by adrenodoxin. J. Biol. Chem. 255 (1980) 3057-3061. 129. Fevold R. H.: Regulation of the adrenal and gonadal 114. Hanukoglu I., Spitsberg V., Bumpus J. A., Dus K. M. microsomal mixed function of steroid and Jefcoate C. R.: Adrenal mitochondrial cytochrome hormone biosynthesis. A. Rev. Physiol. 45 (1983) P-450scc: Cholesterol and adrenodoxin interactions at 19-36. equilibrium and during turnover. J. Biol. Chem. 256 130. Brody R. I. and Black V. H.: Differential ACTH (1981) 4321-4328. response of immanodetectable HMG coenzyme A 115. Greenfield N. J. and Parsons R.: The side-chain cleav- reductase and cytochromes P450-17~ and P450-21 in age of -III. The effect of adrenodoxin, guinea pig adrenal outer zone cell types, zona glomeru- membrane phospholipids and tween 80 on the kinetics losa and zona fasciculata. Endocrine Res. 17 (1991) of oxidation of the sulfate by a reconstituted 195-208. cholesterol desmolase system. J. Steroid Biochem. 24 131. Black V. H.: Cytochrome P450s in the guinea pig (1986) 909-916. adrenal that are immunologically similar to liver forms: 116. Mochizuki H., Suhara K. and Katagiri M.: Adrenal Estrogen suppression explains male-female differences. P-450scc catalyzes deoxycorticosterone 6~-hydroxylase J. Steroid Biochem. Molec. Biol. 43 (1992) 863-868. reaction. J. Steroid Biochem. Molec. Biol. 37 (1990) 132. Provencher P. H., Tremblay Y., Fiet J. and Belanger 133-136. A.: Effect of ACTH on steroidogenic enzymes in 117. Mitani F., Shimizu T., Ueno R., Ishimura Y., Izumi S., guinea pig fasciculata-glomerulosa cells: changes in Komatsu N. and Watanabe K.: Cytochrome P-45011~ activity and mRNA levels. J. Steroid Biochem. Molec. and P-450scc in adrenal cortex: Zonal distribution and Biol. 41 (1992) 59-67. intramitochondrial localization by the horseradish 133. Provencber P. H., Tremblay Y., Caron S. and Belanger peroxidase-labeled antibody method. J. Histochem. A.: Effect of chronic ACTH treatment on guinea-pig Cytoehem. 30 (1982) 1066-1074. adrenal steroidogenesis: steroid plasma levels, steroid 118. Geuze H. J., Slot J. W., Yanagibashi K., McCracken adrenal levels, activity of steroidogenic enzymes and J. A., Schwartz A. L. and Hall P. F.: Immunogold their steady-state mRNA levels. J. Steroid Biochem. cytoehemistry of cytochromes P-450 in porcine Molec. Biol. 41 (1992) 69-78. adrenal cortex. Histochemistry 86 (1987) 551-557. 134. Provencher P. H., Tremblay Y., Belanger B. and 119. Payne A. H.: Hormonal regulation of cytochrome B~langer A.: Steroidogenesis in guinea pig adrenal P450 enzymes, cholesterol side-chain cleavage and 17,,- cortex: effects of ACTH on steroid secretion and hydroxylase/C 17-20 lyase in Leydig cells. Biol. Reprod. steroidogenic enzyme activities and expression. 42 (1990) 399-404. J. Steroid Biochem. Molec. Biol. 43 (1992) 855-862. 120. Akwa Y., Young J., Kabbadi K., Sancho M. J., 135. Hedin L., Rodgers R. J., Simpson E. R. and Richards Zucman D., Vourc'h C., Jung-Testas I., Hu Z. Y., J. S.: Changes in content of cytochrome P45017~, Le Goascogne C., Jo D. H., Corpechot C., Simon P., cytochrome P450scc, and 3-hydroxy-3-methylglutaryl Baulieu E. E. and Robel P.: : Bio- CoA reductase in developing rat ovarian follicles and synthesis metabolism and function of pregnenolone corpora lutea: Correlation with theca cell steroido- and dehydroepiendrosterone in the brain. J. Steroid genesis. Biol. Reprod. 37 (1987) 211-223. Biochem. Molec. Biol. 40 (1991) 71-81. 136. Sasano H., Okamoto M., Mason J. I., Simpson E. R., 121. Hanukoglu I. and Hanukoglu Z.: Stoichiometry of Mendelson C. R., Sasano N. and Silverberg S. G.: mitochondrial cytochromes P-450, adrenodoxin and Immunolocalization of aromatase, 17~-hydroxylase and adrenodoxin reductase in adrenal cortex and corpus side-chain cleavages cytochromes P450 in the human luteurn: Implications for membrane organization and ovary. J. Reprod. Fert. 85 (1989) 163-169. gene regulation. Eur. J. Biochem. 157 (1986) 27-31. 137. Voutilainen R., Tapanainen J., Chung B.-C., Matteson 122. Tuckey R. C., Kostadinovic Z. and Stevenson P. M.: K. J. and Miller W. L.: Hormonal regulation of P450scc Ferredoxin and cytochrome P-450scc concentrations (20,22-desmolase) and P450c17 (17,,-hydroxylase/ in granulosa cells of porcine during follicular 17,20-1yase) in cultured human granulosa cells. J. Clin. cell growth and luteinization. J. Steroid Biochem. 31 Endocr. Metab. 63 (1986) 202-207. (1988) 201-205. 138. Adashi E.: The ovarian life cycle. In Reproductive 123. Simpson E. R. and Miller D. A.: Cholesterol side chain Endocrinology (Edited by S. S. C. Yen and R. B. cleavage cytochrome P450, and iron-sulfur protein in Jaffe). W. B. Saunders, Philadelphia, 4rd Ed (1991) human placental mitochondria. Archs Biochem. Biophys. pp. 181-237. 190 (1978) 800-808. 139. Lashansky G., Saenger P., Fishman K., Gautier T., 124. Walther B., Ghersi-Egea J.-F., Minn A. and Siest G.: Mayes D., Berg G., Di Martino-Nardi J. and Reiter Brain mitochondrial cytochrome P-450scc: spectral E.: Normative data for adrenal steroidogenesis in a and catalytic properties. Archs Biochem. Biophys. 254 healthy pediatric population: Age- and sex-related (1987) 592-596. changes after adrenocorticotropin stimulation. J. Clin. 125. de Launoit Y., Simard J., Durocher F. and Labile F.: Endocr. Metab. 73 (1991) 674-686. Androgenic 17ff-hydroxysteroid dehydrogenase activity 140. Kominami S., Ogawa N., Morimune R., De-Ying H. of expressed rat type I 3]/-hydroxysteroid dehydro- and Takemori S.: The role ofcytochrome b5 in adrenal 800 ISRAEL HANUKOGLU

microsomal steroidogenesis. J. Steroid Biochem. droxygenase (17fl-HSD), and its relation to the ultra- Molec. Biol. 42 (1992) 57-64. structure of stcroidogenic cells in immature and mature 141. Higuchi A., Kominami S. and Takemori S.: Kinetic rat ovaries. Archs Hiatol. Jap. 50 (1987) 545-556. control of steroidogenesis by steroid concentration in 158. Hanukoglu I., Karavolas H. J. and Goy R. W.: guinea pig adrenal mierosomes. Biochim. Biophys Acta Progesterone metabolism in the pineal, brain stem, 1084 (1991) 240-246. thalamus, and corpus callosum of the female rat. Brain 142. Kuhn-Velten W. N., Bunse T. and Forster M. E. C.: Res. 125 (1977) 313-324. Enzyme kinetic and inhibition analyses of cytochrome 159. C¢lotti F., Melcangi R. C., Negri-Cesi P. and Poletti P450XVII, a protein with a bifunctional catalytic site. A.: Testosterone metabolism in brain ceUs and mem- Quantification of effective substrate concentrations at branes. J. Steroid Biochem. Molec. Biol. 40 (1991) the active site and their significance for intrinsic control 673-678. of the hydroxylase/lyase reaction sequence. J. Biol. 160. Lambeth J. D., Scybert D. W., Lancaster J. R., Salerno Chem. 266 (1991) 6291-6301. J. C. and Kamin H.: Steroidogenic electron transport 143. Kitamura M., Buczko E. and Dufau M. L.: Dissoci- in adrenal cortex mitochondria. Molec. Cell. Biochem. ation of hydroxylase and lyase activities by site-directed 45 (1982) 13-31. mutagenesis of the rat P45017a, Molec. Endocr. 5 161. Vermilion J. L., Ballou D. P., Massey V. and Coon (1991) 1373-1380. M. J.: Separate roles for FMN and FAD in catalysis 144. Barnes H. J., Aflotto M. P. and Waterman M. R.: by liver microsomal NADPH-cytochrome P-450 Expression and enzymatic activity of recombinant cyto- reducta~. J. Biol. Chem. 256 (1981) 266-277. chrome P450 17a-liydroxylase in Escherichia coli. 162. Monier S., Van Luc P., Kreibich G., Sabatini D. D. Proc. Natn. Acad. Sci. U.S.A. 88 (1991) 5597-5601. and Adcsnik M.: Signals for the incorporation and 145. Perrin A., Defaye G., Guidicelli C. and Chambaz orientation of cytochrome P450 in the endoplasmic E. M.: Purification and properties of steroid 17~- reticulum membrane. J. Cell. Biol. 107 (1988) 457- hydroxylase from calf testis. Jr. Steroid Biochem. 470. Molec. Biol. 40 (1991) 431-435. 163. Nef P., Heldman J., Lazard D., Margalit T., Jaye M., 146. Sasano H., White P. C., New M. I. and Sasano N.: Hanukoglu I. and Lancet D.: Olfactory-specific cyto- Immunohistochemistry of cytochrome P-450 21- chrome P450; cDNA cloning of a novel neuroepithelial hydroxylase: Microscopic examination of the enzyme enzyme possibly involved in chemoreception. J. Biol. in the bovine adrenal cortex and kidney. Endocrinology Chem. 264 (1989) 6780-6785. 122 (1988) 291-295. 164. Farkash Y., Timberg R. and Orly J.: Preparation of 147. Tukey R. H., Okino S., Barnes H., Gril~n K. J. and antiserum to rat cytochrome P-450 cholesterol side Johnson E. F.: Multiple gene-like sequences related to chain cleavage and its use for ultrastructural localization the rabbit-hepatic progesterone 21-hydroxylase cyto- of the immunoreactive enzyme by protein A-gold chrome P-450 1. J'. Biol. Chem. 260 (1985) 13347-13354. technique. Endocrinology 118 (1986) 1353-1365. 148. John M. E., John M. C., Simpson E. R. and Waterman 165. Eisenberg D., Schwarz E., Komaromy M. and Wall R.: M. R.: Regulation of cytochrome P-45011fl gene Analysis of membrane and surface protein sequences expression by adrenocorticotropin. J. Biol. Chem. 260 with the hydrophobic moment plot. J. Molec. Biol. 179 (1985) 5760-5767. (1984] 125-142. 149. LeHoux J.-G. and Tremblay A.: In vivo regulation 166. Engelman D. M., Steitz T. A. and Goldman A.: of gene expression of enzymes controlling aldosterone Identifying nonpolar transbilayer helices in amino acid synthesis in rat adrenal. J. Steroid Biochem. Molec. sequences of membrane proteins..4. Rev. Biophys. Biol. 43 (1992) 837-846. Chem. 15 (1986) 321-353. 150. Shibata H., Ogishima T., Mitani F., Suzuki H., 167. Hanukoglu I., Suh B. S., Himmelhoch S. and Murakami M., Saruta T. and Ishimura Y.: Regulation Amsterdam A.: Induction and mitochonddal localiz- of aldosterone synthase cytochrome P-450 in rat ation of cytochrome P450scc system enzymes in normal adrenals by angiotensin II and potassium. Endocrinology and transformed ovarian granulosa cells. J. Cell Biol. 128 (1991) 2534-2539. 111 (1990) 1373-1381. 151. Rainey W. E., Bird I. M. and Mason J. I.: Angiotensin- 168. Shiraki H. and Guengerich F. P.: Turnover of mem- II-directed glomerulosa cell function in fetal adrenal brane proteins: kinetics of induction and degradation cells. J. Steroid Biochem. Molec. Biol. 43 (1992) of seven forms of rat liver microsomal cytochrome 847-854. P-450, NADPH-cytochrome /'-450 reductase, and 152. Morohashi K., Nonaka Y., Kiilta S., Hatano O., epoxide ..4rchs Biochem. Biophys. 235 (1984) Takakusu A., Okamoto M. and Omura T.: Enzymatic 86-96. activities of P-450(1 lfl)s expressed by two cDNAs in 169. Furuya S., Okada M., Ito A., Aoyagi H., Kanmera COS-7 cells. J. Biochem. 107 (1990) 635-640. T., Kato T., Sagara Y., Horiuchi T. and Omura T.: 153. Raeside J. I., Renaud R. L. and Friendship R. M.: Synthetic partial extension peptides of P-450(SCC) Isolation of I lfl-liydroxylated androgens from testicular and adrenodoxin precursors: Effects on the import of blood of the mature boar. Biochem. Biophys. Res. mitochondriai enzyme precursors. J. Biochem. 102 Comraun. 162 (1989) 1194-1199. 0987) 821-832. 154. Richards J. S. and Hedin L.: Molecular aspects of 170. Kumamoto T., Morohashi K., Ito A. and Omura T.: hormone action in ovarian follicular development, Site-directed mutagenesis of basic amino acid residues in ovulation, and luteinization. A. Rev. Physiol. 50 (1988) the extension peptide of P-450(SCC) precursor: Effects 441-463. on the import of the precursor into mitoeliondria. 155. Inano H. and Tamaoki B.: Testicular 17fl-hydroxy- J. Biochem. 102 (1987) 833-838. steroid dehydrogenase: Molecular properties and 171. Kumamoto T., Ito A. and Omura T.: Critical region reaction mechanism. Steroids 40 (1986) 3-26. in the extension peptide for the import of cytochrome 156. Luu-The V., Labrie C., Simard J., Lachance Y., P-450(SCC) precursor into mitoehondria. J. Biochem. Zhao H.-F., Couet J., Leblanc G. and Labile F.: 105 (1989) 72-78. Structure of two in tandem human 17fl-hydroxysteroid 172. Matocba M. F. and Waterman M. R.: Synthesis and dehydrogenase genes. Molec. Endocr. 4 (1990) 268-275. processing of mitochondrial steroid hydroxylases. 157. Yoshinaga-Hirabayashi T., Ishimura K., Fujita H., In vivo maturation of the precursor forms of eytochrome Inano H., Ishii-Ohba H. and Tamaoki B.: Immuno- P-450sce, cytochrome P-45011fl, and adrenodoxin. cytochemieal localization of 17fl-hydrosteroid dehy- J. Biol. Chem. 260 (1985) 12259-12265. Steroidogenic enzymes 801

173. Ogishima T., Okada Y. and Omura T.: Import and 190. Purvis J. L., Canick J. A., Latif S. A., Rosenbaum processing of the precursor of cytochrome P-450(scc) J. H., Hologgitas J. and Menard R. H.: Lifetime of by bovine adrenal cortex mitochondria. J. Biochem. 98 microsomal cytochrome P-450 and steroidogenic (1985) 781-791. enzymes in rat testis as influenced by human chorionic 174. Ou W., Ito A., Morohashi K., Fujii-Kuriyama Y. gonadotropin. Archs Biochem. Biophys. 159 (1973) and Omura T.: Processing-independent in vitro trans- 39-49. location of cytochrome P-450(SCC) precursor across 191. Purvis J. L., Canick J. A., Mason J. I., Estabrook mitochondrial membranes. J. Biochem. 100 (1986) R. W. and McCarthy J. L.: Lifetime of adrenal 1287-1296. cytochrome P-450 as influenced by ACTH. Ann. N.Y. 175. Sagara Y., Ito T. and Omura T.: Partial purification of Acad. Sci. 212 (1973) 319-343. a metalloprotease catalyzing the processing of adreno- 192. Boggaram V., John M. E., Simpson E. R. and doxin precursor in bovine adrenal cortex mitochondria. Waterman M. R.: Effect of ACTH on the stability J. Biochem. 96 (1984) 1743-1752. of mRNAs encoding bovine adrenocortical P-450scc, 176. Schatz G.: Signals guiding proteins to their correct P-45011~, P-45017~t, P-450C21 and adrenodoxin. locations in mitochondria. Eur. J. Biochem. 165 (1987) Biochem. Biophys. Res. Commun. 160 (1989) 1227- 1-6. 1232. 177. Ou W. J., Ito A., Umeda M., Inoue K. and Omura T.: 193. Amsterdam A., Rotmensch S., Furman A., Venter Specific binding of mitochondrial protein precursors E. A. and Vlodavsky I.: Synergistic effect of human to liposomes containing cardiolipin. J. Biochem. 103 chorionic gonadotropin and extracellular matrix on (1988) 589-595. in vitro differentiation of human granulosa cells: 178. Matocha M. F. and Waterman M. R.: Discriminatory Progesterone production and gap junction formation. processing of the precursor forms of cytochrome P- Endocrinology 124 (1989) 1956-1964. 450scc and adrenodoxin by adrenocortical and 194. Davoren J. B. and Hsueh A. J. W.: Influence of in vitro mitochondria. J. Biol. Chem. 259 (1984) 8672-8678. plating density on rat granulosa cell responsiveness 179. Akiyoshi-Shibata M., Sakaki T., Yabusaki Y., to follicle-stimulating hormone. Life Sci. 39 (1986) Murakami H. and Ohkawa I-I.: Expression of bovine 1143-I 150. adrenodoxin and NADPH-adrenodoxin reductase 195. Pate J. L., Nephew K. P. and Zarle G. S.: Cell density cDNAs in Saccharomyces cerevisiae. DNA Cell Biol. 10 influences hormonal responsiveness but not lipoprotein (1991) 613-621. utilization in cultured bovine luteal cells. Molec. Cell. 180. Urban P., Breakfield X. O. and Pompon D.: Endocr. 53 (1987) 187-194. Expression of membrane-bound flavoenzymes in yeast. 196. Erickson G. F.: Primary cultures of ovarian cells in In Flavins and 1990 (Edited by B. Curti, serum-free medium as models of hormone-dependent S. Ronchi and G. Zanetti). Walter de Gruyter, Berlin differentiation. Molec. Cell. Endocr. 29 (1983) 21-49. (1991) pp. 869-872. 197. Cauter E. V.: Endocrine rhythms. In Principles and 181. Kalb V. F. and Loper J. C.: Proteins from eight Practice of Endocrinology and Metabolism (Edited by eukaryotic cytochrome P-450 families share a seg- K. L. Becker). J. B. Lippincott, Philadelphia, PA mented region of sequence similarity. Proc. Natn Acad. (1990) pp. 45-52. Sci. U.S.A. 85 (1988) 7221-7225. 198. Avgerinos P. C., Schurmeyer T. H., Gold P. W., 182. Scrutton N. S., Berry A. and Perham R. N.: Redesign Tomai T. P., Loriaux D. L., Sherins R. J., Cutler of the coenzyme specificity of a dehydrogenase by G. B. Jr and Chrousos G. P.: Pulsatile administration protein engineering. Nature 343 (1990) 38-43. of human corticotropin-releasing hormone in patients 183. Peterson J. A., Boddupalli S. S., Ravichandran K. G., with secondary : Restoration of Hasemann C. A., Graham-Lorence S., Tuck S. F., the normal cortisol secretory pattern. J. Clin. Endocr. Ortiz de Montellano P. R. and Deisenhofer J.: Metab. 62 (1986) 816-821. Cytochrome P-450BM-3: A structural and functional 199. James V. H. T.: Adrenal steroid endocrinology--some model for microsomal cytochromes P450. J. Basic unsolved problems. J. Steroid Biochem. Molec. Biol. 39 Clin. Physiol. Pharmac. 3 (Suppl.) (1992) 32-33. (1991) 867-872. 184. Poulos T. L., Finzel B. C. and Howard A. J.: High- 200. Young A. K. and Walker B. L.: The circadian cycles resolution crystal structure of cytochrome P450cam. of plasma corticosterone and adrenal cholesteryl esters J. Molec. Biol. 195 (1987) 687-700. in normal and EFA-deficient female rat. 13 185. Lorence M. C., Trant J. M., Clark B. J., Khyatt B., (1978) 181-186. Mason J. I., Estabrook R. W. and Waterman M. R.: 201. Munck A., Guyre P. M. and Holbrook N. J.: Physio- Construction and expression of human/bovine logical functions of giucocorticoids in stress and their P45017~ chimeric proteins: evidence for distinct tertiary relation to pharmacological actions. Endocrine Rev. 5 structures in the same P450 from different species. (1984) 25-44. Biochemistry 29 (1990) 9819-9824. 202. Bahr V., I-Iense J., Hader O., Bolke T. and Oelkers W.: 186. Sakaki T., Shibata M., Yabnsaki Y., Murakami H. Simulation of steroid secretion by adrenocorticotropin and Ohkawa I-I.: Expression of bovine cytochrome injections and by arginine infusions: no P450c21 and its fused enzymes with yeast NADPH- evidence for a direct stimulation of the human adrenal cytochrome P450 reductase in Saccharomyces by arginine vasopressin. Acta Endocr. 125 (1991) cerevisiae. DNA Cell Biol. 9 (1990) 603-614. 348-353. 187. Hsu L., Hu M. and Chung B.: Structure and function 203. Besedovsky H. O., Del Rey A., Kinsman I., Furukawa of P450c21. J. Basic Clin. Physiol. Pharmac. 3 (Suppl.) H., Monge Arditi G. and Kabiersch A.: Cytokines as (1992) 56-57. modulators of the -pituitary-adrenal 188. Hanukoglu I.: Current research on steroid metabolism: axis. J. Steroid Biochem. Molec. Biol. 40 (1991) transition from biochemistry to molecular-cell biology. 613-618. J. Steroid Biochem. Molec. Biol. 43 (1992) 745-749. 204. Nickels D. A. and Moore D. C.: Serum cortisol 189. Boggaram V., Zuber M. X. and Waterman M. R.: responses in febrile chicken. Pediat. Infect. Dis. J. 8 Turnover of newly synthesized cytochromes P-450scc (1989) 16-19. and P-45011// and adrenodoxin in bovine adreno- 205. Lephart E. D., Baxter C. R. and Parker C. R. Jr.: cortical cells in monolayer culture: Effect of adreno- Effect of burn trauma on adrenal and testicular steroid corticotropin. Archs Biochem. Biophys. 231 (1984) hormone production. J. Clin. Endocr. Metab. 64 (1987) 518-523. 842-848. 802 ISRAEL HANUKOGLU

206. Lambert F., Lammerant J. and Kolanowski J.: steroids during the rat oestrous cycle. Acta Endocr. 85 The prolonged stimulatory effect of ACTH on 1 lfl- (1977) 449-455. hydroxylation, and its contribution to the steroidogenic 223. Zlotkin T., Farkash Y. and Orly J.: Cell-specific potency of adrenocortical cells. J. Steroid Biochem. 20 expression of immunoreactive cholesterol side-chain (1984) 863-868. cleavage cytochrome P-450 during follicular develop- 207. Boshier D. P., Rebuffat P. and Nussdorfer G. G.: ment in the rat ovary. Endocrinology 119 (1986) Cellular responses of the rat adrenal zona fasciculata 2809-2820. to acute ACTH stimulation: A morphometric study. 224. Steinkampf M. P., Mendelson C. R. and Simpson Endocrine Res. 16 (1990) 377-389. E. R.: Regulation by follicle-stimulating hormone of 208. Dallman M. F.: Control of adrenocortical growth the synthesis of aromatase cytochrome P-450 in human in vivo. Endocrine Res. 10 (1985) 213-242. granulosa cells. Molec. Endocr. 1 (1987)465-471. 209. Perkins L. M. and Payne A. H.: Quantification of 225. Rodgers R. J., Waterman M. R. and Simpson E. R.: P-450scc P45017~t, and iron sulfur protein reductase Cytochrome P-450scc, P-45017~, adrenodoxin, and in Leydig cells and adrenals of inbred strains of mice. reduced nicotinamide adenine dinucleotide phosphate- Endocrinology 123 (1988) 2675-2682. cytochrome P-450 reductase in bovine follicles and 210. Lephart E. D., Simpson E. R. and Trzeciak W. H.: corpora lutea. Changes in specific contents during the Rat adrenal 5~t-reductase mRNA content and enzyme ovarian cycle. Endocrinology llg (1986) 1366 1374. activity are dependent. J. Molec. Endocr. 226. Rodgers R. J., Waterman M. R. and Simpson E. R.: 6 (1991) 163-170. Levels of messenger ribonucleic acid encoding chol- 211. Kapas S., Orford C. D., Barker S., Vinson G. P. and esterol cytochrome P-450, 17~-hydroxylase cytochrome Hinson J. P.: Studies on the intraceilular mechanism P-450, adrenodoxin, and low density lipoprotein of action of ~t-melanocyte-stimulating hormone on rat receptor in bovine follicles and corpora lutea through- adrenal zona glomerulosa. J. Molec. Endocr. 9 (1992) out the ovarian cycle. Molec. Endocr. 1 (1987) 47-54. 274-279. 212. Nozu K., Matsuura S., Cart K. J. and Dufau M. L.: 227. Couet J., Martel C., Dupont E., Luu-The V., Sirard Modulation of Leydig cell androgen biosynthesis and M.-A., Zhao H. F., Pelletier G. and Labrie F.: cytochrome P-450 levels during estrogen treatment and Changes in 3fl-hydroxysteroid dehydrogenase/AS-A4 human chorionic gonadotropin-induced desensitization. isomerase messenger ribonucleic acid, activity and J. Biol. Chem. 256 (1981) 10012-10017. protein levels during the estrous cycle in the bovine 213. O'Shaughnessy P. J.: Steroidogenic enzyme activity in ovary. Endocrinology 127 (1990) 2141-2148. the hypogonadal (hpg) mouse testis and effect of treat- 228. Doody K. J., Lephart E. D., Stirling D., Lorence M. ment with . J. Steroid Biochem. C., Magness R. R., McPhaul M. J. and Simpson E. R.: Molec. Biol. 39 (1991) 921-928. Expression of mRNA species encoding steroidogenic 214. Veldhuis J. D.: The hypothalamic-pituitary-testicular enzymes in the rat ovary. J. Molee. Endocr. 6 (1991) axis. In Reproductive Endocrinology (Edited by S. S. C. 153-162. Yen and R. B. Jaffe). W. B. Saunders, Philadelphia, 229. Goldring N. B., Durica J. M., Lifka J., Hedin L., 3rd Edn (1991) pp. 409-459. Ratoosh S. L., Miller W. L., Orly J. and Richards J. S.: 215. Hales D. B., Xiong Y. and Tur-Kaspa I.: The role of Cholesterol side-chain cleavage P450 messenger ribo- cytokines in the regulation of Leydig cell P450c 17 gene nucleic acid: Evidence for hormonal regulation in rat expression. J. Steroid Biochem. Molec. Biol. 43 (1992) ovarian follicles and constitutive expression in corpora 907-914. lutea. Endocrinology 120 (1987) 1942-1950. 216. Orava M., Voutilaninen R. and Vihko R.: Interferon-y 230. Keyes P. L. and Wiltbank M. C.: Endocrine regulation inhibits steroidogenesis and accumulation of mRNA of the corpus luteum. ,4. Rev. Physiol. 50 (1988) of the steroidogenic enzymes P450scc and P450¢17 in 465-482. cultured porcine Leydig cells. Molec. Endocr. 3 (1989) 231. Alila H. W., Dowd J. P., Corradino R. A., Harris 887-894. W. V. and Hansel W.: Control of progesterone pro- 217. Punnonen R., Nummi S., Ylikorkala O., Alapiessa U., duction in small and large bovine luteal cells separated Karvonen P. and Viinikka L.: A composite picture of at flow cytometry. J. Reprod. Fert. 82 (1988) 645-655. the normal menstrual cycle. Acta Obstet. Gynec. Scand. 232. Nelson S. E., McLean M. P., Jayatilak P. G. and (Suppl.) 51 (1975) 65-69. Gibori G.: Isolation, characterization, and culture of 218. Koyama T., de la Pena A. and Hagino N.: Plasma cell subpopulations forming the pregnant rat corpus estrogen, progestin, and luteinizing hormone during luteum. Endocrinology 130 (1992) 954-966. the normal menstrual cycle in the baboon: Role of 233. Voutilainen R. and Miller W. L.: Coordinate tropic luteinizing hormone. Am. J. Obstet. Gynec. 127 (1977) hormone regulation of mRNAs for insulin-like growth 67-72. factor II and the cholesterol side-chain-cleavage 219. Goodman A. L., Desclazi C. D., Johnson D. K. and enzyme P450scc, in human steroidogenic tissues. Hodgen G. D.: Composite pattern of circulating LH, Proc. Nam. Acad. Sci. U.S.A. 84 (1987) 1590-1594. FSH, estradiol, and progesterone during the menstrual 234. Voutilainen R., Picado-Leonard J., DiBlasio A. M. cycle in cynomolgus monkeys. Proc. Soe. Exp. Biol. and Miller W. L.: Hormonal and development regula- Med. 155 (1977) 479-481. tion of adrenodoxin messenger ribonucleic acid in 220. Weiss G., Butler W. R., Hotchkiss J., Dierschke D. J. steroidogenic tissues. J. Clin. Endoer. Metab. 66 (1988) and Knobil E.: Periparturitional serum concentrations 383-388. of , the gonadotropins, and the gonadal 235. Funkenstein B., Waterman M. R. and Simpson E. R.: hormones in the Rhesus monkey. Proc. Soc. Exp. Biol. Induction of synthesis of cholesterol side chain cleav- Med. 151 (1976) 113-116. age cytochrome P-450 and adrenodoxin by follicle- 221. Schams D., Schallenberger E., Hoffmann B. and stimulating hormone, 8-bromo-cyclic AMP, and low Karg H.: The oestrous cycle of the cow: Hormonal density lipoprotein in cultured bovine granulosa cells. parameters and time relationships concerning oestrus, J. Biol. Chem. 2,$9 (1984) 8572-8577. ovulation and electrical resistance of the vaginal mucus. 236. Veldhuis J. D. and Rodgers R. J.: Mechanisms sub- Acta Endoer. 86 (1977) 180-192. serving the steroidogenic synergism between follicle- 222. Kalra P. S. and Kalra S. P.: Temporal changes in the stimulating hormone and insulin-like I hypothalamic and serum luteinizing hormone-releasing (somatomedin C). J. Biol. Chem. 262 (1987) 7658- hormone (LH-RH) levels and the circulating ovarian 7664. Steroidogenic enzymes 803

237. Urban R. J., Garmey J. C., Shupnik M. A. and cytochrome P-450 and adrenodoxin. Archs Biochem. Veldhuis J. D.: Follicle stimulating hormone increases Biophys. 238 (1985) 378-387. concentrations of messenger ribonuclcic acid encoding 252. Roesler W. J., Vandenbark G. R. and Hanson R. W.: cytochrome P450 cholesterol side-chain cleavage Cyclic AMP and the induction of eukaryotic enzyme in primary cultures of porcine granulosa cells. gene transcription. J. Biol. Chem. 263 (1988) Endocrinology 128 (1991) 2000-2007. 9063-9066. 238. Trzeciak W. H., Waterman M. R. and Simpson E. R.: 253. Hatey F., Gasparoux J. P., Mulsant P., Bonnet A. Synthesis of the cholesterol side-chain cleavage enzymes and Gasser F.: P450scc regulation in pig granulosa in cultured rat ovarian granulosa cells: Induction by cells: investigation into the mechanism of induction. follicle-stimulating hormone and dibutyryl adenosine J. Steroid Biochem. Molec. Biol. 43 (1992) 869-874. Y,5'-monophosphate. Endocrinology 119 (1986) 323- 254. Hales D. B., Sha L. and Payne A. H.: Glucocorticoid 330. and cyclic adenosine 3'5'-monophosphatc-mediatcd 238a. Tilly J. L., Kowalski K. I. and Johnson A. L.: induction of cholesterol side-chain cleavage cytochrome Cytochrome P450 side-chain cleavage (P450scc) in the P450 (P450scc) in MA-10 tumor Lcydig cells. Increases hen ovary. II. P450scc messenger RNA, immuno- in mRNA are cycloheximide sensitive. Endocrinology reactive protein, and enzyme activity in developing 126 (1990) 2800-2808. granulosa cells. Biol. Reprod. 45, (1991) 967-974. 255. Golos T. G., Miller W. L. and Strauss III J. F.: Human 239. Ben-Ze'ev A., Kohen F. and Amsterdam A.: Gonado- chorionic gonadotropin and 8-bromo cyclic adenosine tropin-induced differentiation of granulosa cells is monophosphate promote an acute increase in cyto- associated with the co-ordinated regulation of cyto- chrome P450scc and adrenodoxin messenger RNAs in skeletal proteins involved in cell-contact formation. cultured human granulosa cells by a cycloheximide- Differentiation 34 (1987) 222-235. insensitive mechanism. J. Clin. Invest. 80 (1988) 240. Amsterdam A., Hanukoglu I., Sub B. S., Keren-Tal I., 869-899. Plehn-Dujowich D., Springel R., Rennert H. and 256. Mellon S. H. and Valsse C.: cAMP regulates P450scc Strauss J. F. III: Oncogene-transformed granulosa ~ne expression by a cycloheximide insensitive mechan- cells as a model system for the study of steroidogenic ism in cultured mouse Leydig MA-10 cells. Proc. Natn. processes. J. Steroid Biochem. Molec. Biol. 43 (1992) Acad. Sci. U.S.A. 86 (1990) 7775-7779. 875-884. 257. Viard I., Hall S. H., Jalllard C., Bertheion M. C. and 241. Goldschmit D., Kraicer P. and Orly J,: Periovulatory Saez J. M.: Regulation of C-Fos, C-Jun and Jun-B expression of cholesterol side-chain cleavage cyto- messenger ribonucleic acids by angiotensin-II and chrome P-450 in cumulus cells. Endocrinology 124 corticotropin in ovine and bovine adrenocortical cells. (1989) 369-378. Endocrinology 130 (1992) 1193-1200. 242. Amsterdam A., Koch Y., Liebcrman M. E. and 258. McMahon S. B. and Monroe J. G.: Role of primary Lindner H. R.: Distribution of binding sites for human response genes in generating cellular responses to chorionic gonadotropin in the preovulatory follicle of growth factors. FASEB J. 6 (1992) 2707-2715. the rat. J. Cell Biol. 67 (1975) 894-900. 259. Cohen P.: Signal integration at the level of protein 243. Lawrence T. S., Dekel N. and Beers W. H.: Binding kinases, protein phosphatases and their substrates. of human chorionic gonadotropin by rat cumuli Trends Biochem. Sci. 17 (1992) 408-414. oophori and granulosa cells: A comparative study. 260. Liscovitch M.: Crosstalk among multiple signal- Endocrinology 106 (1980) 1114-1118. activated phospbolipases. Trends Biochem. Sci. 17 244. Tsafriri A.: Local regulators of ovarian (1992) 393-398. function. In The Physiology of (Edited 261. Mooibrock M. and Wang J. H.: Integration of signal- by E. Knobil and J. Neill). Raven Press, New York transduction processes. Biochem. Cell Biol. 66 (1988) (1988) pp. 527-565. 557-566. 245. Hammond J. M., Mondschein J. S., Samaras S. E. and 262. Nishizuka Y.: Signal transduction: crosstalk. Trends Canning S. F.: The ovarian insulin-like growth factors, Biochem. Sci. 17 (1992) 367. a local amplification mechanism for steroidogenesis 263. Pazin M. L and Williams L. T.: Triggering signaling and hormone action. J. Steroid Biochem. Molec. Biol. cascades by receptor tyrosine kinases. Trends Biochem. 40 (1991) 411-416. Sci. 17 (1992) 374-378. 246. Constantino C. X., Keyes P. L. and Kostyo J. L.: 263a. Sato T., Sakaguchi M., Mihara K. and Omura T.: Insulin-like growth factor-1 stimulates steroidogenesis The amino terminal structures that determine topolog- in rabbit luteal cells. Endocrinology 128 (1991) 1702- ical orientation of cytochrome P-450 in microsomal 1708. membrane. EMBO Jl 9 (1990) 2391-2397. 247. Trzeciak W. H., Waterman M. R., Simpson E. R. and 264. Kerlavage A. R.: G-protein-coupled receptor family. Ojeda S. R.: Vasoactive intestinal pcptide regulates Curr. Opin. Struc. Biol. 1 (1991) 394-401. cholesterol side-chain cleavage cytochrome P-450 265. Mountjoy K. G., Robbins L. S., Mortrud M. T. and (P-450scc) gene expression in ganulosa cells from Cone R. D.: The cloning of a family of genes that immature rat ovaries. Molec. Endocr. 1 (1987) 500- encode the melanocortin receptors. Science 257 (1992) 504. 1248-1251. 248. Mori M. and Marsh J. M.: The site of luteinizing 266. Hepler J. R. and Gilman A. G.: G proteins. Trends hormone stimulation of steroidogenesis in mitochon- Biochem. Sci. 17 (1992) 383-387. dria of the rat corpus luteum. J. Biol. Chem. 257 (1982) 267. Bollag W. B., Barrett P. Q., Isales C. M. and 6178-6183. Rasmussen H.: Angiotensin II-induced changes in di- 249. Holt J. A.: Regulation of progesterone production acylglycerol levels and their potential role in modulating in the rabbit corpus luteum. Biol. Reprod. 40 (1989) the steroidogenic response. Endocrinology 128 (1991) 201-208. 231-241. 250. Waterman M. R. and Simpson E. R.: Regulation of 268. Cooke B. A., Dirami G., Chaudry L., Choi M. S. K., steroid hydoxylase gene expression is multifactorial in Abayasekara D. R. E. and Phipp L.: Release of nature. Recent Prog. Horm. Res. 45 (1989) 533-568. arachidonic acid and the effects of 251. Anderson C. M. and Mendeison C. R.: Regulation of on steroidogenesis in rat testis Leydig cells. J. Steroid steroidogenesis in rat Lcydig cells in culture: Effect of Biochem. Molec. Biol. 40 (1991) 465-471. human chorionic gonadotropin and dibutyryl cyclic 269. Farese R. V.: Phosphoinositide metabolism and AMP on the synthesis of cholesterol side chain cleavage hormone action. Endocrine Rev. 4 (1983) 78-95. 804 ISP,Ar~ HANUKOGLU

270. Farese R. V. and Prudente W. J.: Localization of the kinase in the expression of cholesterol side chain metabolic processes affected by calcium during cortico- cleavage and steroid 1l~-hydroxylase genes in mouse tropin action. Biochim. Biophys. Acta 497 (1977) adrenocortical tumor cells. J. Biol. Chem. 264 (1989) 386-395. 12867-12871. 271. Flores J. A., Veldhuis J. D. and Leong D. A.: Follicle- 283. Woody C. J. and LaBarbera A. R.: Adenylyl cyclase stimulating hormone evokes an increase in intracellular of perifused porcine granulosa cells remains responsive free calcium ion concentrations in single ovarian to pulsatile, but not continuous stimulation with fol- (granulosa) cells. Endocrinology 127 (1990) 3172- licle-stimulating hormone. Endocrinology 124 (1989) 3179. 2261- 2267. 272. Fukuda N., Honda M. and Hatano M.: Role of cal- 284. Naseeruddin S. A. and Hornsby P. J.: Regulation of cium ion in ACTH-induced steroidogenesis in humans. 11~- and 17,~-hydroxylases in cultured bovine adreno- J. Steroid Biochem. 31 (1988) 337-344. cortical cells: 3',5'-cyclic adenosine monophosphate, 273. Ganguly A., Chiou S., West L. and Davis J. S.: Pro- insulin like growth factor-I, and activators of protein opiomelanocortin-derived peptides phosphoinositides. kinase C. Endocrinology 127 (1990) 1673-1681. cAMP and aldosterone secretion. J. Steroid Biochem. 285. Chang C. W., Naseeruddin S. A. and Hornsby P. J.: 33 (1989) 1143-1148. The response of 21-hydroxylase mRNA levels to cyclic 274. Garren L. D., Gill G. N., Masui H. and Walton G. M.: AMP and TPA in bovine adrenocortical cells is On the mechanism of action of ACTH. Recent Prog. dependent on culture conditions. Endocrinology 128 Horm. Res. 27 (1971) 433-474. (1991) 604-610. 275. Gudermann T., Birnbaumer M. and Birnbaumer L.: 286. Gomberg-Malool S., Ziv R., Posner I., Levitzki A. and Evidence for dual coupling of the murine luteinizing Orly J.: Tyrphostins inhibit FSH mediated functions hormone re~ptor to adenyl cyclase and phosphoinosit- in cultured rat ovarian granulosa cells. Endocrinology ide breakdown and Ca 2+ mobilization. Studies with the (1992) in press. cloned murine luteinizing expressed 287. Widmaier E. P. and Hall P. F.: Protein kinase C in in L cells. J. Biol. Chem. 267 (1992) 4479-4488. adrenal cells: possible role in regulation of steroid 276. Mattioli M., Barboni B. and Scren E.: Luteinizing synthesis. Molec. Cell. Endocr. 43 (1985) 181-188. hormone inhibits potasssium outward currents in swine 288. Hoyer P. B. and Kong W.: Protein kinase A and C granulosa cells by intracellular calcium mobilization. activities and endogenous substrates in ovine small Endocrinolgy 129 (1991) 2740-2745. and large luteal cells. Molec. Cell. Endocr. 62 (1989) 277. Morley P., Schwartz J. L., Whitfield J. F. and Tsang 203-215. B. D.: Role of chloride ions in progesterone production 289. Brentano S. T., Picardo-Lconard J., Melion S. H., by chicken granulosa cells. Molec. Cell. Endocr. 82 Moore C. C. D. and Miller W. L.: Tissue specific, (1991) 107-115. cyclic adenosine Y-5'-monophosphate-induced, and 278. Neuman I., Solano A. R., Paz C., Mele P., phorbol ester-repressed transcription from the human Maciel F. C., Lemos J. R., Fernandez H. N. and P450c17 promoter in mouse cells. Molec. Endocr. 4 Podesta E. J.: The action of luteinizing hormone on (1.990) 1972-1979. the testis. J. Steroid Biochem. Molec. Biol. 40 (1991) 290. Suh B. S. and Amsterdam A.: Establishment of highly 441-451. steroidogenic granulosacell lines by cotransfection with 279. Ramachandran J.: Corticotropin receptors, cyclic SV40 and Ha-ras oncogene: Induction of steroido- AMP and steroidogenesis. Endocrine Res. 10 (1985) genesis by cyclic adenosine 3'-5'-monophosphate and 347-363. its suppression by phorbol ester. Endocrinology 127 280. Solano A. R., Dada L. and Podesta E. J.: Lipoxy- (1990) 2489-2500. genase products as common intermediates in cyclic 291. Meduri G., Vuhai-Luuthi T., Jolivet A. and AMP-dependent and independent adrenal steroido- Milgrom E.: New functional zonation in the ovary genesis in rats. J. Molec. Endocr. I (1988) 14%154. as shown by immunohistochemistry of luteinizing 281. Steele G. L. and Leung P. C.: Intragonadal signalling hormone receptor. Endocrinology 131 (1992) 366-373. mechanisms in the control of steroid hormone pro- 292. Camp T. A., Rahal J. O. and Mayo K. E.: Cellular duction. J. Steroid Biochem. Molec. Biol. 41 (1992) localization and hormonal regulation of follicle-stimu- 515-522. lating hormone and luteinizing hormone receptor mes- 282. Wong M., Rice D. A., Parker K. L. and Schimmer senger RNAs in the rat ovary. Molec. Endocr. 5 (1991) B. P.: The role of cAMP and cAMP-dependent protein 1405-1417.