Open Access
How to translate text using browser tools
1 August 2015 Regulation of Steroidogenesis, Development, and Cell Differentiation by Steroidogenic Factor-1 and Liver Receptor Homolog-1
Takashi Yazawa, Yoshitaka Imamichi, Kaoru Miyamoto, Md. Rafiqul Islam Khan, Junsuke Uwada, Akihiro Umezawa, Takanobu Taniguchi
Author Affiliations +
Abstract

Steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1) belong to the nuclear receptor superfamily and are categorized as orphan receptors. In addition to other nuclear receptors, these play roles in various physiological phenomena by regulating the transcription of target genes. Both factors share very similar structures and exhibit common functions. Of these, the roles of SF-1 and LRH-1 in steroidogenesis are the most important, especially that of SF-1, which was originally discovered and named to reflect such roles. SF-1 and LRH-1 are essential for steroid hormone production in gonads and adrenal glands through the regulation of various steroidogenesis-related genes. As SF-1 is also necessary for the development of gonads and adrenal glands, it is also considered a master regulator of steroidogenesis. Recent studies have clearly demonstrated that LRH-1 also represents another master regulator of steroidogenesis, which similarly to SF-1, can induce differentiation of non-steroidogenic stem cells into steroidogenic cells. Here, we review the functions of both factors in these steroidogenesis-related phenomena.

INTRODUCTION

Nuclear receptors (NRs) belong to a large superfamily of transcription factors, which are essential for various physiological phenomena (Mangelsdorf et al., 1995; Robinson-Rechavi et al., 2003). They are activated in a lipophilic ligand-dependent manner in response to stimulation by steroid hormones, thyroid hormones, vitamins, and various lipids. In addition to such typical classes, NRs include orphan receptors whose ligands have not been identified. Some orphan NRs are constitutively activated in a ligand-independent fashion; steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1) are two such receptors and are members of the NR5A subfamily (SF-1 is NR5A1, and LRH-1 is NR5A2), together with Drosophila FTZ-F1 (NR5A3) (Fayard et al., 2004; Lee and Moore, 2008; Parker and Schimmer, 1997; Schimmer and White, 2010) (Fig. 1A). Both transcription factors play important roles in controlling the endocrine functions of various tissues. Of these, the regulation of steroidogenesis is one of the most important roles for both factors. In particular, SF-1 was originally identified as a transcription factor that is essential for the transcription of steroidogenic enzyme genes.

Adrenal glands and gonads are the primary steroidogenic organs in mammals and other amniotes (Miller, 1988; Miller and Auchus, 2011). Organ-derived steroid hormones are involved in various physiological phenomena. Adrenal steroids (glucocorticoid and mineralocorticoid) are essential for glucose metabolism, stress responses, immunity, and fluid/electrolyte balance. Gonadal sex steroids (androgens and estrogens) are important for sex differentiation and reproduction. Ovarian progesterone is necessary for ovulation and pregnancy. Steroid hormones are synthesized from cholesterol, which is delivered to the inner compartments of mitochondria by steroidogenic acute regulatory protein (StAR) (Papadopoulos and Miller, 2012; Stocco, 1997, 2000). Cholesterol is converted to pregnenolone by P450 side chain cleavage enzyme (P450scc/CYP11A1/Cyp11a1), a rate-limiting enzyme in the synthesis of all steroid hormones (Miller, 2008; Papadopoulos and Miller, 2012). Thereafter, tissue-specific P450 hydroxylases and hydroxysteroid dehydrogenases catalyze reactions that produce tissue-specific steroid hormones (Miller, 1988, 2008; Miller and Auchus, 2011).

It is well known that SF-1 and LRH-1 are involved in the transcription of steroidogenesis-related genes (Fayard et al., 2004; Lee and Moore, 2008; Parker and Schimmer, 1997; Schimmer and White, 2010; Wang et al., 2001; Yazawa et al., 2014; Yazawa et al., 2011) and that steroidogenesis is markedly influenced by a deficiency of either factor (Duggavathi et al., 2008; Jeyasuria et al., 2004; Pelusi et al., 2008; Zhang et al., 2013). SF-1 is also known to be involved in the development of steroidogenic organs (Luo et al., 1994; Parker and Schimmer, 1997) and the differentiation of steroidogenic cells (Yazawa et al., 2014; Yazawa et al., 2009; Yazawa et al., 2006). For these reasons, SF-1 has long been known as a master regulator of steroidogenesis. More recently, a number of groups, including ours, have shown that LRH-1 is another master regulator for steroidogenesis (Bouguen et al., 2015; Yazawa et al., 2009; Yazawa et al., 2011; Zhang et al., 2013). The present review outlines the discovery of SF-1 and LRH-1 and their roles in steroidogenesis-related phenomena and their functional differences.

Fig. 1.

Schematic structure of SF-1 and LRH-1. DBD, Hinge and LBD represent DNA-binding domain, hinge region and ligand-binding domain, respectively. Black boxes of C-terminus show an AF-2 motif. The percentages indicate the levels of amino-acid identity between each region of human proteins. Ovarian LRH-1 is different from liver form only in N-terminal domain (white boxes).

f01_323.jpg

Table 1.

Target genes of SF-1 and LRH-1, which are involved in steroidogenesis.

t01_323.gif

General characteristics of SF-1 and LRH-1

Early studies demonstrated that the promoter regions of multiple cytochrome P450 steroid hydroxylase genes include an AGGTCA-like motif (Ad4 sequence) that is essential for their transcription in steroidogenic cells (Honda et al., 1990; Rice et al., 1991). Keith Parker and Ken Morohashi independently identified a transcription factor in mouse and bovine adrenal glands that binds to this motif (Lala et al., 1992; Morohashi et al., 1992). Because this transcription factor confers promoter activity to P450 steroid hydroxylase genes in non-steroidogenic cells, they respectively named it steroidogenic factor-1 (SF-1)/adrenal 4-binding protein (Ad4BP). Consistent with this role, detection of SF-1/Ad4BP in adults revealed its expression in the three layers of the adrenal cortex (zona reticularis, zona fasciculata, and zona glomerulosa), testicular Leydig and Sertoli cells, ovarian theca, granulosa cells (GCs), and to a lesser extent in the corpus lutea (Hatano et al., 1994; Ikeda et al., 1993; Kawabe et al., 1999). From these findings Parker and Morohashi concluded that SF-1/Ad4BP is a determining factor for cell-specific expression of cytochrome P450 steroid hydroxylase genes. Consistent with this hypothesis, SF-1/Ad4BP regulates the transcription of all cytochrome P450 steroid hydroxylase genes (Carlone and Richards, 1997; Guo et al., 2003; Hanley et al., 2001; Hu et al., 2001; Lala et al., 1992; Lin et al., 2001; Lynch et al., 1993; Michael et al., 1995; Morohashi et al., 1993; Schimmer and Parker, 1992; Wang et al., 2000; Ye et al., 2009; Zhang and Mellon, 1996) (Table 1). Additionally, the transcription of various steroidogenesis-related genes is regulated by SF-1/Ad4BP, such as cholesterol deliverers (Cao et al., 1997; Caron et al., 1997; Sugawara et al., 1996), hydroxysteroid dehydrogenase (Leers-Sucheta et al., 1997), and electron transporters (Huang et al., 2005; Imamichi et al., 2013, 2014) (Table 1).

LRH-1 was first discovered in mouse liver as a transcription factor that regulates the transcription of α-fetoprotein (Becker-Andre et al., 1993; Galarneau et al., 1996). LRH-1 functions in the control of glycolysis as well as cholesterol and bile acid homeostasis by regulating the transcription of various genes, such as glucokinase (Oosterveer et al., 2012), SR-BI (Schoonjans et al., 2002), CYP7A1, and CYP8B1 (Goodwin et al., 2000; Lu et al., 2000). In addition to liver, LRH-1 is highly expressed in tissues of endodermal origin (pancreas and intestine), and involved in metabolism, inflammation, and stem cell renewal (Benod et al., 2011; Botrugno et al., 2004; Coste et al., 2007; Fayard et al., 2003; Fernandez-Marcos et al., 2011; Lazarus et al., 2012). LRH-1 is also expressed in testicular Leydig cells and ovarian GCs, and regulates the transcription of steroidogenesis-related genes (Peng et al., 2003; Pezzi et al., 2004; Yazawa et al., 2009; Yazawa et al., 2010). In particular, expression levels of LRH-1 are more abundant in ovary than in any other tissue (Bookout et al., 2006; Kanno et al., 2014). Recently, it was shown that the ovarian LRH-1 represents a unique isoform, which has a truncated N-terminal domain (Kawabe et al., 2013) (Fig. 1). Although the functional difference between both isoforms is not clear, their expression is controlled by completely distinctive transcriptional regulation (Kawabe et al., 2013; Zhang et al., 2001). LRH-1 can replace SF-1 in progesterone production of the corpus luteum (CL) to maintain pregnancy (Zhang et al., 2013). It is also expressed in embryonic stem (ES) cells and regulates the expression of central self-renewal factors, Oct-3/4 and Nanog (Gu et al., 2005; Mullen et al., 2007; Wagner et al., 2010). Interestingly, LRH-1 enhances the efficiency of reprograming somatic cells into induced pluripotent stem (iPS) cells (Guo and Smith, 2010; Heng et al., 2010; Wang et al., 2011).

SF-1/Ad4BP and LRH-1 belong to the same NR subfamily and share many structural and functional characteristics. Both proteins contain the structural domains of typical NRs, a zinc finger DNA-binding domain, intervening hinge region, and a carboxyl-terminal putative ligand-binding domain, although SF-1 lacks the N-terminal domain containing an AF-1 motif (Fig. 1). It has been proposed that the long hinge region exhibits AF-1-like activation activity (Hammer et al., 1999; Li et al., 1999). Because their DNA-binding domains are particularly similar (Fig. 1), SF-1 and LRH-1 effectively recognize the same DNA sequence, YCAAGGYCR (Y represents a pyrimidine, and R represents a purine), in regulatory elements of target genes (Fayard et al., 2003). Both factors thus commonly regulate the transcription of the same genes. This phenomenon is also applicable to steroidogenesis-related genes, including cytochrome P450 steroid hydroxylases (Kim et al., 2005; Pezzi et al., 2004; Wang et al., 2001; Yazawa et al., 2009; Yazawa et al., 2010; Yazawa et al., 2011), cholesterol deliverers (Kim et al., 2004; Schoonjans et al., 2002), hydroxysteroid dehydrogenase (Peng et al., 2003) and electron transporter (Imamichi et al., 2013) (Table 1). Thus, in addition to SF-1, it is conceivable that LRH-1 also works as a determining factor in cell-specific expression of steroidogenesis-related genes. In support of this notion, SF-1 and LRH-1 share common functions in other phenomena, such as transcriptional regulation of Oct-3/4 (Barnea and Bergman, 2000; Gu et al., 2005) and the generation of iPS cells (Guo and Smith, 2010; Heng et al., 2010).

Inveolvement of SF-1 and LRH-1 in the development and steroidogenesis of gonads and adrenal glands

The requirement for SF-1 in the development of steroidogenic organs has been demonstrated in vivo using various gene-targeted models. SF-1-knockout (KO) mice die of glucocorticoid deficiency shortly after birth, and exhibit male-to-female sex reversal in external genitalia (Luo et al., 1994; Sadovsky et al., 1995). These phenotypes are caused by the complete loss of gonads and adrenal glands. Although gonadal and adrenal development initiate without SF-1, they regress via apoptotic cell death during the subsequent developmental stage, possibly due to an abnormality in glycolysis and the pentose phosphate pathway (Baba et al., 2014). In SF-1-KO mice, because the gonads disappear prior to the production of anti-Müllerian hormone and testosterone, which are necessary for the induction of male sexual differentiation, the internal and external urogenital tracts are of the female type, even in genetic males. Heterozygous KO mice also show a decrease in adrenal gland volume, which is associated with an impairment of corticosterone production in response to stress (Bland et al., 2004; Bland et al., 2000; Fatchiyah et al., 2006). These KO mouse models demonstrate that SF-1 functions as a master regulator in the development of steroidogenic organs in vivo. Consistent with the phenotypes of KO mice, patients with mutations in the SF-1 gene exhibit gonadal and adrenal defects (Achermann et al., 1999; Ferraz-de-Souza et al., 2011; Hasegawa et al., 2004; Lourenco et al., 2009). In addition to steroidogenic organs, SF-1 gene mutations and KO result in defects of the pituitary gonadotroph (Ingraham et al., 1994; Shinoda et al., 1995; Zhao et al., 2001), ventromedial hypothalamus (Ikeda et al., 1995; Shinoda et al., 1995; Zhao et al., 2008), and spleen (Morohashi et al., 1999; Zangen et al., 2014).

In tissue-specific KO models, it has been shown that SF-1 plays important roles in steroidogenesis and other homeostatic processes following organ development. In Leydig cell-specific KO mice, there are marked decreases in testicular Star and Cyp11a1 expression, indicating a defect in testosterone production (Jeyasuria et al., 2004). Consistent with this hypothesis, the testes fail to descend (an androgen-dependent developmental process) and are hypoplastic. In GC-specific KO mice, there are numerous ovarian defects (Pelusi et al., 2008). In these mice, the ovaries are hypoplastic, adults are sterile, and ovaries show reduced numbers of oocytes. Gonadotropin-induced estrogen and progesterone production are also markedly reduced in this model. These findings show that SF-1 expression is essential in steroidogenic organs throughout life.

Total ablation of LRH-1 causes a severe reduction of Oct-3/4 expression levels and an embryonic lethal phenotype at around embryonic day 6.5–7.5 (Gu et al., 2005; Pare et al., 2004). The functions of LRH-1 in vivo have additionally been revealed by examination of heterozygous and tissue-specific KO models. In heterozygous Lrh-1 KO mice, the abnormalities of gonadal steroidogenesis are exhibited in both males and females (Labelle-Dumais et al., 2007; Volle et al., 2007). In males, testicular testosterone production is decreased by a reduction in the expression of steroidogenesis-related genes such as Star, Cyp11a1, and Hsd3b (Volle et al., 2007). Accordingly, the epididymides and seminal vesicles of these mice have smaller weights than those of wild-type mice. In females, there is a reduction in the progesterone production of luteal cells, resulting in a subfertile phenotype (Labelle-Dumais et al., 2007). The importance of LRH-1 in female reproduction was further strengthened by Murphy and colleagues using GC- and CL-KO models (Bertolin et al., 2014; Duggavathi et al., 2008; Zhang et al., 2013). GC-KO mice are infertile because of anovulation and impairment of progesterone production due to failure of luteinization (Bertolin et al., 2014; Duggavathi et al., 2008). However, even though CL-KO mice ovulate normally, they are infertile because of implantation failure caused by a deficiency in progesterone production (Zhang et al., 2013). Thus, in addition to SF-1, LRH-1 is also necessary for steroidogenesis in some tissues during postnatal life.

Differentiation of stem cells into steroidogenic cells by SF-1 and LRH-1

In support of the phenotypes of gene-targeted models, we have demonstrated that SF-1 and LRH-1 induce differentiation of non-steroidogenic stem cells into steroidogenic cells (Miyamoto et al., 2011; Yazawa et al., 2014; Yazawa et al., 2009; Yazawa et al., 2010; Yazawa et al., 2011; Yazawa et al., 2006; Yazawa et al., 2008) (Fig. 2). Among the various types of stem cells, we focused on mesenchymal stem cells (MSCs). MSCs are multipotent somatic stem cells that originate in the mesoderm, as is the case with steroidogenic organs. They are defined as adherent fibroblast-like cells that can differentiate into osteoblasts, adipocytes, and chondrocytes (Fig. 2), although they can also generate cells of all three germ layers, at least in vitro (Gojo and Umezawa, 2003; Prockop, 1997; Toyoda et al., 2007). MSCs were originally isolated from bone marrow (BM-MSCs) by Friedenstein et al. (Friedenstein et al., 1976), and have been identified in fat, placenta, umbilical cord blood, and other tissues (Hass et al., 2011; Kode et al., 2009). Because they may be a source of connective tissue lineages in vivo, it is plausible that MSCs are present in most organs throughout the body.

Fig. 2.

Induction of gonadal and adrenal steroidogenic cells from multipotent MSCs. With aid of cAMP, both SF-1 and LRH-1 independently have a capacity to differentiate MSCs into steroidogenic cells.

f02_323.jpg

To explore the potential to differentiate into steroidogenic cells, rat BM-MSCs were transplanted into immature testes at the same stage that adult Leydig cells begin to differentiate from stem/progenitor mesenchymal cells (Yazawa et al., 2006). After several weeks, transplanted MSCs had colonized the interstitial spaces of recipient testes and expressed Leydig cell markers, such as Cyp11a1, Hsd3b1, and Cyp17a1. Using a promoter-sorting approach with the human CYP11A1 promoter (genome sequences that regulate cell-specific expression of the CYP11A1 gene in gonadal and adrenal steroidogenic cells), we demonstrated that some isolated mouse BM-MSCs spontaneously differentiate into cells expressing steroidogenic enzymes (Yazawa et al., 2006). These results indicate that MSCs have a capacity to differentiate into steroidogenic cells both in vivo and in vitro. In addition, stable transfection of SF-1 and cAMP treatment induces differentiation of all mouse MSCs into Cyp11a1-positive steroidogenic cells. These cells express various steroidogenic enzymes, and produce progesterone and testosterone. This method can also differentiate human BM-MSCs into cortisol-producing cells in response to adrenocorticotropic hormone stimulation, which are similar to the zona fasciculata cells of the adrenal gland. Additionally, BM-MSCs transform to steroidogenic cells by adenovirus-mediated transient expression of SF-1 (Mizutani et al., 2010; Tanaka et al., 2007; Wei et al., 2012; Yanase et al., 2006). These studies are consistent with the concept that SF-1 is a master regulator for steroidogenesis. In addition to SF-1, introduction of LRH-1 also efficiently induces the differentiation of BM-MSCs into steroidogenic cells without SF-1 (Yazawa et al., 2009; Yazawa et al., 2011). In LRH-1-itroduced cells, expression levels of steroidogenic enzymes and steroid hormone productions were comparable with those in SF-1-introduced cells, suggesting that LRH-1 could be another master regulator in steroidogenesis. In fact, such situations likely occur in CL and intestinal epithelial cells (IECs). These cells synthesize progesterone or glucocorticoid, and although the expression of SF-1 is nearly undetectable (Mueller et al., 2007; Peng et al., 2003; Ramayya et al., 1997), LRH-1 is highly expressed (Mueller et al., 2007; Peng et al., 2003). It is thus conceivable that steroidogenesis of CL and lECs is depend solely on LRH-1, a hypothesis that is supported by the disordered steroidogenesis of these cells in conditional KO mice (Coste et al., 2007; Zhang et al., 2013).

As is the case for BM-MSCs, MSCs derived from other tissues can be differentiated into steroidogenic cells using the above methods (Gondo et al., 2008; Wei et al., 2012; Yazawa et al., 2014; Yazawa et al., 2010). However, these methods are not applicable to pluripotent stem cells, such as ES cells, iPS cells, and embryonal carcinoma cells, because SF-1 and LRH-1 are cytotoxic to these cells (Yazawa et al., 2011; Yazawa et al., 2006). In an early study, Crawford et al., also reported that even though ectopic expression of SF-1 only rarely induces ES cells to express the Cyp11a1 gene, these cells do not synthesize steroid hormone due to the lack of cholesterol transporter (Crawford et al., 1997; Yazawa et al., 2006). These phenomena are likely because of the properties of LRH-1 that are important for Oct-3/4 expression in these cells (Gu et al., 2005). Oct-3/4 expression levels must be constant in order to maintain the undifferentiated status of pluripotent stem cells (Niwa et al., 2000). Overexpression of LRH-1 (or ectopic expression of SF-1) can markedly affect the expression levels of Oct-3/4 (Gu et al., 2005; Heng et al., 2010). Therefore, it is reasonable to conclude that these factors cause difficulties for the direct induction of steroidogenic cells from pluripotent stem cells. However, mouse ES cells do differentiate into adrenal cortex-like cells when they are pre-differentiated into MSCs by pulse treatment with retinoic acid before expressing SF-1 (Yazawa et al., 2011). These results indicate that MSCs are very useful tools for inducing steroidogenic cells using NR5A subfamily proteins. Our results also suggest that pluripotent stem cells can differentiate into steroidogenic cells by these factors after pre-differentiation Into the mesodermal lineage. Consistent with this hypothesis, It was demonstrated by Sonoyama et al. that human ES and IPS cells can differentiate Into cortisol-producing cells by the expression of SF-1 only after pre-differentiation Into mesodermal cells (Sonoyama et al., 2012). In addition, Jameson and colleague reported that pre-selection or EB formation is necessary to differentiate SF-1-introduced mouse ES cells into sex steroid-producing cells (Jadhav and Jameson, 2011).

CONCLUSION

NR5A subfamily proteins are essential for various phenomena associated with steroidogenesis. SF-1 and LRH-1 activate the transcription of steroidogenesis-related genes by binding to the same DNA sequences of their regulatory regions. SF-1 is a master regulator for the development of gonads and adrenal glands as well as for steroidogenesis following organogenesis. Conversely, LRH-1 is unlikely to be involved in the development of these tissues, although it has been shown to be important for gonadal steroidogenesis. Both factors induce the differentiation of non-steroldogenic stem cells into steroidogenic cells. It is thus clear that SF-1 and LRH-1 have common functions in steroidogenesis, although future studies should resolve the differences between them. For example, as mentioned above, the localization and expression levels of SF-1 and LRH-1 are quite different in steroidogenic tissues. Additionally, although both factors are expressed in Leydig cells and GCs simultaneously, a deficiency of either factor cannot be compensated by another factor. These findings suggest that both factors have unique functions. In GCs, it is noteworthy that ovarian estrogen production is reduced by repression of aromatase expression in SF-1 GC-KO mice (Pelusi et al., 2008), but it is unaffected in LRH-1 GC-KO mice (Duggavathl et al., 2008). Further studies are necessary to reveal the functions of SF-1 and LRH-1 in steroidogenesis.

ACKNOWLEDGMENTS

We thank Ms. S. Tsunoda for administrative support. This work was supported in part by JSPS KAKENHI Grant Number 23590329 (Grant-in-Aid for Scientific Research (C)), 25460378 (Grant-in-Aid for Scientific Research (O)) and 26860170 (Grant-in-Aid for Young Scientists (B)) granted by Japan Society for the Promotion of Science, the Smoking Research Foundation, and the fund for Asahikawa Medical University Creative Research in the Field of Life Science.

REFERENCES

1.

JC Achermann , M Ito , M Ito , PC Hindmarsh , JL Jameson ( 1999) A mutation in the gene encoding steroidogenic factor-1 causes XY sex reversal and adrenal failure in humans. Nat Genet 22: 125–126 Google Scholar

2.

T Baba , H Otake , T Sato , K Miyabayashi , Y Shishido , CY Wang , et al. ( 2014) Glycolytic genes are targets of the nuclear receptor Ad4BP/SF-1. Nat Commun 5: 3634 Google Scholar

3.

E Barnea , Y Bergman ( 2000) Synergy of SF1 and RAR in activation of Oct-3/4 promoter. J Biol Chem 275: 6608–6619 Google Scholar

4.

M Becker-Andre , E Andre , JF DeLamarter ( 1993) Identification of nuclear receptor mRNAs by RT-PCR amplification of conserved zinc-finger motif sequences. Biochem Biophys Res Commun 194: 1371–1379 Google Scholar

5.

C Benod , MV Vinogradova , N Jouravel , GE Kim , RJ Fletterick , EP Sablin ( 2011) Nuclear receptor liver receptor homologue 1 (LRH-1) regulates pancreatic cancer cell growth and proliferation. Proc Natl Acad Sci USA 108: 16927–16931 Google Scholar

6.

K Bertolin , J Gossen , K Schoonjans , BD Murphy ( 2014) The orphan nuclear receptor Nr5a2 is essential for luteinization in the female mouse ovary. Endocrinology 155: 1931–1943 Google Scholar

7.

ML Bland , CA Jamieson , SF Akana , SR Bornstein , G Eisenhofer , MF Dallman , HA Ingraham ( 2000) Haploinsufficiency of steroidogenic factor-1 in mice disrupts adrenal development leading to an impaired stress response. Proc Natl Acad Sci USA 97: 14488–14493 Google Scholar

8.

ML Bland , RC Fowkes , HA Ingraham ( 2004) Differential requirement for steroidogenic factor-1 gene dosage in adrenal development versus endocrine function. Mol Endocrinol 18: 941–952 Google Scholar

9.

AL Bookout , Y Jeong , M Downes , RT Yu , RM Evans , DJ Mangelsdorf ( 2006) Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 126: 789–799 Google Scholar

10.

OA Botrugno , E Fayard , JS Annicotte , C Haby , T Brennan , O Wendling , et al. ( 2004) Synergy between LRH-1 and betacatenin induces G1 cyclin-mediated cell proliferation. Mol Cell 15: 499–509 Google Scholar

11.

G Bouguen , L Dubuquoy , P Desreumaux , T Brunner , B Bertin (2015) Intestinal steroidogenesis. Steroids Google Scholar

12.

G Cao , CK Garcia , KL Wyne , RA Schultz , KL Parker , HH Hobbs ( 1997) Structure and localization of the human gene encoding SR-BI/CLA-1. Evidence for transcriptional control by steroidogenic factor 1. J Biol Chem 272: 33068–33076 Google Scholar

13.

DL Carlone , JS Richards ( 1997) Functional interactions, phosphorylation, and levels of 3′,5′-cyclic adenosine monophosphate-regulatory element binding protein and steroidogenic factor-1 mediate hormone-regulated and constitutive expression of aromatase in gonadal cells. Mol Endocrinol 11: 292–304 Google Scholar

14.

KM Caron , Y Ikeda , SC Soo , DM Stocco , KL Parker , BJ Clark ( 1997) Characterization of the promoter region of the mouse gene encoding the steroidogenic acute regulatory protein. Mol Endocrinol 11: 138–147 Google Scholar

15.

A Coste , L Dubuquoy , R Barnouin , JS Annicotte , B Magnier , M Notti , et al. ( 2007) LRH-1-mediated glucocorticoid synthesis in enterocytes protects against inflammatory bowel disease. Proc Natl Acad Sci USA 104: 13098–13103 Google Scholar

16.

PA Crawford , Y Sadovsky , J Milbrandt ( 1997) Nuclear receptor steroidogenic factor 1 directs embryonic stem cells toward the steroidogenic lineage. Mol Cell Biol 17: 3997–4006 Google Scholar

17.

R Duggavathi , DH Volle , C Mataki , MC Antal , N Messaddeq , J Auwerx , et al. ( 2008) Liver receptor homolog 1 is essential for ovulation. Genes Dev 22: 1871–1876 Google Scholar

18.

Fatehiyah , M Zuhair , Y Shima , S Oka , S Ishihara , Y Fukui-Katoh , K Morohashi ( 2006) Differential gene dosage effects of Ad4BP/SF-1 on target tissue development. Biochem Biophys Res Commun 341: 1036–1045 Google Scholar

19.

E Fayard , K Schoonjans , JS Annicotte , J Auwerx ( 2003) Liver receptor homolog 1 controls the expression of carboxyl ester lipase. J Biol Chem 278: 35725–35731 Google Scholar

20.

E Fayard , J Auwerx , K Schoonjans ( 2004) LRH-1: an orphan nuclear receptor involved in development, metabolism and steroidogenesis. Trends Cell Biol 14: 250–260 Google Scholar

21.

PJ Fernandez-Marcos , J Auwerx , K Schoonjans ( 2011) Emerging actions of the nuclear receptor LRH-1 in the gut. Biochim Biophys Acta 1812: 947–955 Google Scholar

22.

B Ferraz-de-Souza , L Lin , JC Achermann ( 2011) Steroidogenic factor-1 (SF-1, NR5A1) and human disease. Mol Cell Endocrinol 336: 198–205 Google Scholar

23.

AJ Friedenstein , JF Gorskaja , NN Kulagina ( 1976) Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol 4: 267–274 Google Scholar

24.

L Galarneau , JF Pare , D Allard , D Hamel , L Levesque , JD Tugwood , et al. ( 1996) The alpha1-fetoprotein locus is activated by a nuclear receptor of the Drosophila FTZ-F1 family. Mol Cell Biol 16: 3853–3865 Google Scholar

25.

S Gojo , A Umezawa ( 2003) Plasticity of mesenchymal stem cells--regenerative medicine for diseased hearts. Hum Cell 16: 23–30 Google Scholar

26.

S Gondo , T Okabe , T Tanaka , H Morinaga , M Nomura , R Takayanagi , et al. ( 2008) Adipose tissue-derived and bone marrow-derived mesenchymal cells develop into different lineage of steroidogenic cells by forced expression of steroidogenic factor 1. Endocrinology 149: 4717–4725 Google Scholar

27.

B Goodwin , SA Jones , RR Price , MA Watson , DD McKee , LB Moore , et al. ( 2000) A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 6: 517–526 Google Scholar

28.

P Gu , B Goodwin , AC Chung , X Xu , DA Wheeler , RR Price , et al. ( 2005) Orphan nuclear receptor LRH-1 is required to maintain Oct4 expression at the epiblast stage of embryonic development. Mol Cell Biol 25: 3492–3505 Google Scholar

29.

G Guo , A Smith ( 2010) A genome-wide screen in EpiSCs identifies Nr5a nuclear receptors as potent inducers of ground state pluripotency. Development 137: 3185–3192 Google Scholar

30.

IC Guo , MC Hu , BC Chung ( 2003) Transcriptional regulation of CYP11A1. J Biomed Sci 10: 593–598 Google Scholar

31.

GD Hammer , I Krylova , Y Zhang , BD Darimont , K Simpson , NL Weigel , HA Ingraham ( 1999) Phosphorylation of the nuclear receptor SF-1 modulates cofactor recruitment: integration of hormone signaling in reproduction and stress. Mol Cell 3: 521–526 Google Scholar

32.

NA Hanley , WE Rainey , DI Wilson , SG Ball , KL Parker ( 2001) Expression profiles of SF-1, DAX1, and CYP17 in the human fetal adrenal gland: potential interactions in gene regulation. Mol Endocrinol 15: 57–68 Google Scholar

33.

T Hasegawa , M Fukami , N Sato , N Katsumata , G Sasaki , K Fukutani , et al. ( 2004) Testicular dysgenesis without adrenal insufficiency in a 46,XY patient with a heterozygous inactive mutation of steroidogenic factor-1. J Clin Endocrinol Metab 89: 5930–5935 Google Scholar

34.

R Hass , C Kasper , S Bohm , R Jacobs ( 2011) Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 9: 12 Google Scholar

35.

O Hatano , K Takayama , T Imai , MR Waterman , A Takakusu , T Omura , K Morohashi ( 1994) Sex-dependent expression of a transcription factor, Ad4BP, regulating steroidogenic P-450 genes in the gonads during prenatal and postnatal rat development. Development 120: 2787–2797 Google Scholar

36.

JC Heng , B Feng , J Han , J Jiang , P Kraus , JH Ng , et al. ( 2010) The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell 6: 167–174 Google Scholar

37.

S Honda , K Morohashi , T Omura ( 1990) Novel cAMP regulatory elements in the promoter region of bovine P-450(11 beta) gene. J Biochem 108: 1042–1049 Google Scholar

38.

MC Hu , NC Hsu , CI Pai , CK Wang , B Chung ( 2001) Functions of the upstream and proximal steroidogenic factor 1 (SF-1)-binding sites in the CYP11A1 promoter in basal transcription and hormonal response. Mol Endocrinol 15: 812–818 Google Scholar

39.

N Huang , A Dardis , WL Miller ( 2005) Regulation of cytochrome b5 gene transcription by Sp3, GATA-6, and steroidogenic factor 1 in human adrenal NCI-H295A cells. Mol Endocrinol 19: 2020–2034 Google Scholar

40.

Y Ikeda , DS Lala , X Luo , E Kim , MP Moisan , KL Parker ( 1993) Characterization of the mouse FTZ-F1 gene, which encodes a key regulator of steroid hydroxylase gene expression. Mol Endocrinol 7: 852–860 Google Scholar

41.

Y Ikeda , X Luo , R Abbud , JH Nilson , KL Parker ( 1995) The nuclear receptor steroidogenic factor 1 is essential for the formation of the ventromedial hypothalamic nucleus. Mol Endocrinol 9: 478–486 Google Scholar

42.

Y Imamichi , T Mizutani , Y Ju , T Matsumura , S Kawabe , M Kanno , et al. ( 2013) Transcriptional regulation of human ferredoxin 1 in ovarian granulosa cells. Mol Cell Endocrinol 370: 1–10 Google Scholar

43.

Y Imamichi , T Mizutani , Y Ju , T Matsumura , S Kawabe , M Kanno , et al. ( 2014) Transcriptional regulation of human ferredoxin reductase through an intronic enhancer in steroidogenic cells. Biochim Biophys Acta 1839: 33–42 Google Scholar

44.

HA Ingraham , DS Lala , Y Ikeda , X Luo , WH Shen , MW Nachtigal , et al. ( 1994) The nuclear receptor steroidogenic factor 1 acts at multiple levels of the reproductive axis. Genes Dev 8: 2302–2312 Google Scholar

45.

U Jadhav , JL Jameson ( 2011) Steroidogenic factor-1 (SF-1)-driven differentiation of murine embryonic stem (ES) cells into a gonadal lineage. Endocrinology 152: 2870–2882 Google Scholar

46.

P Jeyasuria , Y Ikeda , SP Jamin , L Zhao , DG De Rooij , AP Themmen , et al. ( 2004) Cell-specific knockout of steroidogenic factor 1 reveals its essential roles in gonadal function. Mol Endocrinol 18: 1610–1619 Google Scholar

47.

M Kanno , T Yazawa , S Kawabe , Y Imamichi , Y Usami , Y Ju , et al. ( 2014) Sex-determining region Y-box 2 and GA-binding proteins regulate the transcription of liver receptor homolog-1 in early embryonic cells. Biochim Biophys Acta 1839: 406–414 Google Scholar

48.

K Kawabe , T Shikayama , H Tsuboi , S Oka , K Oba , T Yanase , et al. ( 1999) Dax-1 as one of the target genes of Ad4BP/SF-1. Mol Endocrinol 13: 1267–1284 Google Scholar

49.

S Kawabe , T Yazawa , M Kanno , Y Usami , T Mizutani , Y Imamichi , et al. ( 2013) A novel isoform of liver receptor homolog-1 is regulated by steroidogenic factor-1 and the specificity protein family in ovarian granulosa cells. Endocrinology 154: 1648–1660 Google Scholar

50.

JW Kim , N Peng , WE Rainey , BR Carr , GR Attia ( 2004) Liver receptor homolog-1 regulates the expression of steroidogenic acute regulatory protein in human granulosa cells. J Clin Endocrinol Metab 89: 3042–3047 Google Scholar

51.

JW Kim , JC Havelock , BR Carr , GR Attia ( 2005) The orphan nuclear receptor, liver receptor homolog-1, regulates cholesterol side-chain cleavage cytochrome p450 enzyme in human granulosa cells. J Clin Endocrinol Metab 90: 1678–1685 Google Scholar

52.

JA Kode , S Mukherjee , MV Joglekar , AA Hardikar ( 2009) Mesenchymal stem cells: immunobiology and role in immunomodulation and tissue regeneration. Cytotherapy 11: 377–391 Google Scholar

53.

C Labelle-Dumais , JF Pare , L Belanger , R Farookhi , D Dufort ( 2007) Impaired progesterone production in Nr5a2+/- mice leads to a reduction in female reproductive function. Biol Reprod 77: 217–225 Google Scholar

54.

DS Lala , DA Rice , KL Parker ( 1992) Steroidogenic factor I, a key regulator of steroidogenic enzyme expression, is the mouse homolog of fushi tarazu-factor I. Mol Endocrinol 6: 1249–1258 Google Scholar

55.

KA Lazarus , D Wijayakumara , AL Chand , ER Simpson , CD Clyne ( 2012) Therapeutic potential of Liver Receptor Homolog-1 modulators. J Steroid Biochem Mol Biol 130: 138–146 Google Scholar

56.

YK Lee , DD Moore ( 2008) Liver receptor homolog-1, an emerging metabolic modulator. Front Biosci 13: 5950–5958 Google Scholar

57.

S Leers-Sucheta , K Morohashi , JI Mason , MH Melner ( 1997) Synergistic activation of the human type II 3beta-hydroxysteroid dehydrogenase/delta5-delta4 isomerase promoter by the transcription factor steroidogenic factor-1/adrenal 4-binding protein and phorbol ester. J Biol Chem 272: 7960–7967 Google Scholar

58.

LA Li , EF Chiang , JC Chen , NC Hsu , YJ Chen , BC Chung ( 1999) Function of steroidogenic factor 1 domains in nuclear localization, transactivation, and interaction with transcription factor TFIIB and c-Jun. Mol Endocrinol 13: 1588–1598 Google Scholar

59.

CJ Lin , JW Martens , WL Miller ( 2001) NF-1C, Sp1, and Sp3 are essential for transcription of the human gene for P450c17 (steroid 17alpha-hydroxylase/17,20 lyase) in human adrenal NCI-H295A cells. Mol Endocrinol 15: 1277–1293 Google Scholar

60.

D Lourenco , R Brauner , L Lin , A De Perdigo , G Weryha , M Muresan , et al. ( 2009) Mutations in NR5A1 associated with ovarian insufficiency. N Engl J Med 360: 1200–1210 Google Scholar

61.

TT Lu , M Makishima , JJ Repa , K Schoonjans , TA Kerr , J Auwerx , DJ Mangelsdorf ( 2000) Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 6: 507–515 Google Scholar

62.

X Luo , Y Ikeda , KL Parker ( 1994) A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell 77: 481–490 Google Scholar

63.

JP Lynch , DS Lala , JJ Peluso , W Luo , KL Parker , BA White ( 1993) Steroidogenic factor 1, an orphan nuclear receptor, regulates the expression of the rat aromatase gene in gonadal tissues. Mol Endocrinol 7: 776–786 Google Scholar

64.

DJ Mangelsdorf , C Thummel , M Beato , P Herrlich , G Schutz , K Umesono , et al. ( 1995) The nuclear receptor superfamily: the second decade. Cell 83: 835–839 Google Scholar

65.

MD Michael , MW Kilgore , K Morohashi , ER Simpson ( 1995) Ad4BP/SF-1 regulates cyclic AMP-induced transcription from the proximal promoter (PII) of the human aromatase P450 (CYP19) gene in the ovary. J Biol Chem 270: 13561–13566 Google Scholar

66.

WL Miller ( 1988) Molecular biology of steroid hormone synthesis. Endocr Rev 9: 295–318 Google Scholar

67.

WL Miller ( 2008) Steroidogenic enzymes. Endocr Dev 13: 1–18 Google Scholar

68.

WL Miller , RJ Auchus ( 2011) The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev 32: 81–151 Google Scholar

69.

K Miyamoto , T Yazawa , T Mizutani , Y Imamichi , SY Kawabe , M Kanno , et al. ( 2011) Stem cell differentiation into steroidogenic cell lineages by NR5A family. Mol Cell Endocrinol 336: 123–126 Google Scholar

70.

T Mizutani , T Yazawa , Y Ju , Y Imamichi , M Uesaka , Y Inaoka , et al. ( 2010) Identification of a novel distal control region upstream of the human steroidogenic acute regulatory protein (StAR) gene that participates in SF-1-dependent chromatin architecture. J Biol Chem 285: 28240–28251 Google Scholar

71.

K Morohashi , S Honda , Y Inomata , H Handa , T Omura ( 1992) A common trans-acting factor, Ad4-binding protein, to the promoters of steroidogenic P-450s. J Biol Chem 267: 17913–17919 Google Scholar

72.

K Morohashi , UM Zanger , S Honda , M Hara , MR Waterman , T Omura ( 1993) Activation of CYP11A and CYP11B gene promoters by the steroidogenic cell-specific transcription factor, Ad4BP. Mol Endocrinol 7: 1196–1204 Google Scholar

73.

K Morohashi , H Tsuboi-Asai , S Matsushita , M Suda , M Nakashima , H Sasano , et al. ( 1999) Structural and functional abnormalities in the spleen of an mFtz-F1 gene-disrupted mouse. Blood 93: 1586–1594 Google Scholar

74.

M Mueller , A Atanasov , I Cima , N Corazza , K Schoonjans , T Brunner ( 2007) Differential regulation of glucocorticoid synthesis in murine intestinal epithelial versus adrenocortical cell lines. Endocrinology 148: 1445–1453 Google Scholar

75.

EM Mullen , P Gu , AJ Cooney ( 2007) Nuclear Receptors in Regulation of Mouse ES Cell Pluripotency and Differentiation. PPAR Res 2007: 61563 Google Scholar

76.

H Niwa , J Miyazaki , AG Smith ( 2000) Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 24: 372–376 Google Scholar

77.

MH Oosterveer , C Mataki , H Yamamoto , T Harach , N Moullan , TH van Dijk , et al. ( 2012) LRH-1-dependent glucose sensing determines intermediary metabolism in liver. J Clin Invest 122: 2817–2826 Google Scholar

78.

V Papadopoulos , WL Miller ( 2012) Role of mitochondria in steroidogenesis. Best Pract Res Clin Endocrinol Metab 26: 771–790 Google Scholar

79.

JF Pare , D Malenfant , C Courtemanche , M Jacob-Wagner , S Roy , D Allard , L Belanger ( 2004) The fetoprotein transcription factor (FTF) gene is essential to embryogenesis and cholesterol homeostasis and is regulated by a DR4 element. J Biol Chem 279: 21206–21216 Google Scholar

80.

KL Parker , BP Schimmer ( 1997) Steroidogenic factor 1: a key determinant of endocrine development and function. Endocr Rev 18: 361–377 Google Scholar

81.

C Pelusi , Y Ikeda , M Zubair , KL Parker ( 2008) Impaired follicle development and Infertility in female mice lacking steroidogenic factor 1 in ovarian granulosa cells. Biol Reprod 79: 1074–1083 Google Scholar

82.

N Peng , JW Kim , WE Rainey , BR Carr , GR Attia ( 2003) The role of the orphan nuclear receptor, liver receptor homologue-1, in the regulation of human corpus luteum 3beta-hydroxysteroid dehydrogenase type II. J Clin Endocrinol Metab 88: 6020–6028 Google Scholar

83.

V Pezzi , R Sirianni , A Chimento , M Maggiolini , S Bourguiba , C Delalande , et al. ( 2004) Differential expression of steroidogenic factor-1/adrenal 4 binding protein and liver receptor homolog-1 (LRH-1)/fetoprotein transcription factor in the rat testis: LRH-1 as a potential regulator of testicular aromatase expression. Endocrinology 145: 2186–2196 Google Scholar

84.

DJ Prockop ( 1997) Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 276: 71–74 Google Scholar

85.

MS Ramayya , J Zhou , T Kino , JH Segars , CA Bondy , GP Chrousos ( 1997) Steroidogenic factor 1 messenger ribonucleic acid expression in steroidogenic and nonsteroidogenic human tissues: Northern blot and in situ hybridization studies. J Clin Endocrinol Metab 82: 1799–1806 Google Scholar

86.

DA Rice , AR Mouw , AM Bogerd , KL Parker ( 1991) A shared promoter element regulates the expression of three steroidogenic enzymes. Mol Endocrinol 5: 1552–1561 Google Scholar

87.

M Robinson-Rechavi , H Escriva Garcia , V Laudet ( 2003) The nuclear receptor superfamily. J Cell Sci 116: 585–586 Google Scholar

88.

Y Sadovsky , PA Crawford , KG Woodson , JA Polish , MA Clements , LM Tourtellotte , et al. ( 1995) Mice deficient in the orphan receptor steroidogenic factor 1 lack adrenal glands and gonads but express P450 side-chain-cleavage enzyme in the placenta and have normal embryonic serum levels of corticosteroids. Proc Natl Acad Sci USA 92: 10939–10943 Google Scholar

89.

BP Schimmer , KL Parker ( 1992) Promoter elements of the mouse 21-hydroxylase (Cyp-21) gene involved in cell-selective and cAMP-dependent gene expression. J Steroid Biochem Mol Biol 43: 937–950 Google Scholar

90.

BP Schimmer , PC White ( 2010) Minireview: steroidogenic factor 1: its roles in differentiation, development, and disease. Mol Endocrinol 24: 1322–1337 Google Scholar

91.

K Schoonjans , JS Annicotte , T Huby , OA Botrugno , E Fayard , Y Ueda , et al. ( 2002) Liver receptor homolog 1 controls the expression of the scavenger receptor class B type I. EMBO Rep 3: 1181–1187 Google Scholar

92.

K Shinoda , H Lei , H Yoshii , M Nomura , M Nagano , H Shiba , et al. ( 1995) Developmental defects of the ventromedial hypothalamic nucleus and pituitary gonadotroph in the Ftz-F1 disrupted mice. Dev Dyn 204: 22–29 Google Scholar

93.

T Sonoyama , M Sone , K Honda , D Taura , K Kojima , M Inuzuka , et al. ( 2012) Differentiation of human embryonic stem cells and human induced pluripotent stem cells into steroid-producing cells. Endocrinology 153: 4336–4345 Google Scholar

94.

DM Stocco ( 1997) A StAR search: implications in controlling steroidgenesis. Biol Reprod 56: 328–336 Google Scholar

95.

DM Stocco ( 2000) The role of the StAR protein in steroidogenesis: challenges for the future. J Endocrinol 164: 247–253 Google Scholar

96.

T Sugawara , JA Holt , M Kiriakidou , JF Strauss 3rd ( 1996) Steroidogenic factor 1-dependent promoter activity of the human steroidogenic acute regulatory protein (StAR) gene. Biochemistry 35: 9052–9059 Google Scholar

97.

T Tanaka , S Gondo , T Okabe , K Ohe , H Shirohzu , H Morinaga , et al. ( 2007) Steroidogenic factor 1/adrenal 4 binding protein transforms human bone marrow mesenchymal cells into steroidogenic cells. J Mol Endocrinol 39: 343–350 Google Scholar

98.

M Toyoda , H Takahashi , A Umezawa ( 2007) Ways for a mesenchymal stem cell to live on its own: maintaining an undifferentiated state ex vivo. Int J Hematol 86: 1–4 Google Scholar

99.

DH Volle , R Duggavathi , BC Magnier , SM Houten , CL Cummins , JM Lobaccaro , et al. ( 2007) The small heterodimer partner is a gonadal gatekeeper of sexual maturation in male mice. Genes Dev 21: 303–315 Google Scholar

100.

RT Wagner , X Xu , F Yi , BJ Merrill , AJ Cooney ( 2010) Canonical Wnt/beta-catenin regulation of liver receptor homolog-1 mediates pluripotency gene expression. Stem Cells 28: 1794–1804 Google Scholar

101.

XL Wang , M Bassett , Y Zhang , S Yin , C Clyne , PC White , WE Rainey ( 2000) Transcriptional regulation of human 11 beta-hydroxylase (hCYP11B1). Endocrinology 141: 3587–3594 Google Scholar

102.

ZN Wang , M Bassett , WE Rainey ( 2001) Liver receptor homologue-1 1 expressed in the adrenal and can regulate transcription of 11 beta-hydroxylase. J Mol Endocrinol 27: 255–258 Google Scholar

103.

W Wang , J Yang , H Liu , D Lu , X Chen , Z Zenonos , et al. ( 2011) Rapid and efficient reprogramming of somatic cells to induced pluripotent stem cells by retinoic acid receptor gamma and liver receptor homolog 1. Proc Natl Acad Sci USA 108: 18283–18288 Google Scholar

104.

X Wei , G Peng , S Zheng , X Wu ( 2012) Differentiation of umbilical cord mesenchymal stem cells into steroidogenic cells in comparison to bone marrow mesenchymal stem cells. Cell Prolif 45: 101–110 Google Scholar

105.

T Yanase , S Gondo , T Okabe , T Tanaka , H Shirohzu , W Fan , et al. ( 2006) Differentiation and regeneration of adrenal tissues: An initial step toward regeneration therapy for steroid insufficiency. Endocr J 53: 449–459 Google Scholar

106.

T Yazawa , T Mizutani , K Yamada , H Kawata , T Sekiguchi , M Yoshino , et al. ( 2006) Differentiation of adult stem cells derived from bone marrow stroma into Leydig or adrenocortical cells. Endocrinology 147: 4104–4111 Google Scholar

107.

T Yazawa , M Uesaka , Y Inaoka , T Mizutani , T Sekiguchi , T Kajitani , et al. ( 2008) Cyp11b1 is induced in the murine gonad by luteinizing hormone/human chorionic gonadotropin and involved in the production of 11-ketotestosterone, a major fish androgen: conservation and evolution of the androgen metabolic pathway. Endocrinology 149: 1786–1792 Google Scholar

108.

T Yazawa , Y Inanoka , T Mizutani , M Kuribayashi , A Umezawa , K Miyamoto ( 2009) Liver receptor homolog-1 regulates the transcription of steroidogenic enzymes and induces the differentiation of mesenchymal stem cells into steroidogenic cells. Endocrinology 150: 3885–3893 Google Scholar

109.

T Yazawa , Y Inaoka , R Okada , T Mizutani , Y Yamazaki , Y Usami , et al. ( 2010) PPAR-gamma coactivator-1 alpha regulates progesterone production in ovarian granulosa cells with SF-1 and LRH-1. Mol Endocrinol 24: 485–496 Google Scholar

110.

T Yazawa , S Kawabe , Y Inaoka , R Okada , T Mizutani , Y Imamichi , et al. ( 2011) Differentiation of mesenchymal stem cells and embryonic stem cells into steroidogenic cells using steroidogenic factor-1 and liver receptor homolog-1. Mol Cell Endocrinol 336: 127–132 Google Scholar

111.

T Yazawa , Y Imamichi , K Miyamoto , A Umezawa , T Taniguchi ( 2014) Differentiation of mesenchymal stem cells into gonad and adrenal steroidogenic cells. World J Stem Cells 6: 203–212 Google Scholar

112.

P Ye , Y Nakamura , E Lalli , WE Rainey ( 2009) Differential effects of high and low steroidogenic factor-1 expression on CYP11B2 expression and aldosterone production in adrenocortical cells. Endocrinology 150: 1303–1309 Google Scholar

113.

D Zangen , Y Kaufman , E Banne , A Weinberg-Shukron , A Abulibdeh , BP Garfinkel , et al. ( 2014) Testicular differentiation factor SF-1 is required for human spleen development. J Clin Invest 124: 2071–2075 Google Scholar

114.

P Zhang , SH Mellon ( 1996) The orphan nuclear receptor steroidogenic factor-1 regulates the cyclic adenosine 3′,5′-monophosphate-mediated transcriptional activation of rat cytochrome P450C17 (17 alpha-hydroxylase/c17-20 lyase). Mol Endocrinol 10: 147–158 Google Scholar

115.

CK Zhang , W Lin , YN Cai , PL Xu , H Dong , M Li , et al. ( 2001) Characterization of the genomic structure and tissue-specific promoter of the human nuclear receptor NR5A2 (hB1F) gene. Gene 273: 239–249 Google Scholar

116.

C Zhang , MJ Large , R Duggavathi , FJ DeMayo , JP Lydon , K Schoonjans , et al. ( 2013) Liver receptor homolog-1 is essential for pregnancy. Nat Med 19: 1061–1066 Google Scholar

117.

L Zhao , M Bakke , Y Krimkevich , LJ Cushman , AF Parlow , SA Camper , KL Parker ( 2001) Steroidogenic factor 1 (SF1) is essential for pituitary gonadotrope function. Development 128: 147–154 Google Scholar

118.

L Zhao , KW Kim , Y Ikeda , KK Anderson , L Beck , S Chase , et al. ( 2008) Central nervous system-specific knockout of steroidogenic factor 1 results in increased anxiety-like behavior. Mol Endocrinol 22: 1403–1415 Google Scholar
© 2015 Zoological Society of Japan
Takashi Yazawa, Yoshitaka Imamichi, Kaoru Miyamoto, Md. Rafiqul Islam Khan, Junsuke Uwada, Akihiro Umezawa, and Takanobu Taniguchi "Regulation of Steroidogenesis, Development, and Cell Differentiation by Steroidogenic Factor-1 and Liver Receptor Homolog-1," Zoological Science 32(4), 323-330, (1 August 2015). https://doi.org/10.2108/zs140237
Received: 25 October 2014; Accepted: 3 March 2015; Published: 1 August 2015
KEYWORDS
adrenal
gonad
liver receptor homolog-1
steroidogenesis
steroidogenic factor-1
transcriptional regulation
Back to Top