Quick viewing(Text Mode)

Downloaded from Bioscientifica.Com at 09/29/2021 03:48:41AM Via Free Access

Downloaded from Bioscientifica.Com at 09/29/2021 03:48:41AM Via Free Access

62 1

Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R47–R64 development REVIEW The molecular pathways underlying early gonadal development

Yisheng Yang, Stephanie Workman and Megan J Wilson

Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand

Correspondence should be addressed to M J Wilson: [email protected]

Abstract

The body of knowledge surrounding reproductive development spans the fields of Key Words genetics, anatomy, physiology and biomedicine, to build a comprehensive understanding ff gonad of the later stages of reproductive development in and animal models. Despite ff urogenital ridge this, there remains much to learn about the bi-potential progenitor structure that the ff disorders of and testis arise from, known as the genital ridge (GR). This tissue forms relatively determination late in and has the potential to form either the ovary or testis, ff infertility which in turn produce hormones required for the development of the rest of the ff molecular genetics reproductive tract. It is imperative that we understand the genetic networks underpinning GR development if we are to begin to understand abnormalities in the adult. This is particularly relevant in the contexts of disorders of sex development (DSDs) and infertility, two conditions that many individuals struggle with worldwide, with often no answers as to their aetiology. Here, we review what is known about the genetics of GR development. Investigating the genetic networks required for GR formation will not only contribute to our understanding of the genetic regulation of reproductive development, it may in turn open new avenues of investigation into reproductive abnormalities and later fertility Journal of Molecular issues in the adult. Endocrinology (2019) 62, R47–R64

The distinction between is one of the most obvious testicular/ovarian structures, accessory sex structures and examples of dimorphism in the animal kingdom and secondary sexual characteristics), whereby genital ridge highlights one of the most crucial fate decisions made in (GR) formation was included only as a step in gonadal sex utero to develop as a male or a . This fate decision determination or excluded from the broader picture all to follow either developmental trajectory is an essential together. The focus of this review is formation of the GR process that determines the reproductive success of all in mice and humans, and consequences of failure of its sexually reproducing animals (Fig. 1). There are a number development, rather than sex determination, which has of different mechanisms underlying sex development been reviewed extensively elsewhere (Greenfield 2015, that can be broadly categorized as either genetically Capel 2017). sex determined or environmentally determined (Capel 2017). Typically, fall within the genetically sex- Genital ridge formation determined category. Traditionally, sexual development has been viewed as two distinct processes, gonadal sex The mammalian GR is derived from intermediate meso­ determination (decision of the gonad to become a testis derm as paired structures that lie on either side of the dorsal or ovary) and (establishment of mesentery in the coelomic cavity (Fig. 2). Formation of

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access

-17-0314 Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R48 Endocrinology development

2010). As these cells delaminate from the coelomic epithelium, they undergo an epithelial-to-mesenchymal transition (EMT) (Kusaka et al. 2010) and as mesenchymal cells, begin to populate the space between the coelomic epithelium and the (Karl & Capel 1998, Schmahl et al. 2000). These mesenchymal cells of the GR are precursor cells that can differentiate into somatic support cells and interstitial/stromal cell lineages of the early sexually differentiated gonad (Karl & Capel 1998, Ito et al. 2006, Mork et al. 2012). In the testis, additional cells are also recruited into the GR region from the mesonephros to augment the population of mesenchymal cells. Cell-tracing and culture studies in mice revealed that these largely contribute Figure 1 Summary of the origin of reproductive tissues within the mammalian to the endothelial cell population for the establishment of . The gonad region of the GR develops into either the testis or the the male vascular system (Karl & Capel 1995, Martineau ovary depending upon expression of the Sry . In XY gonads, the et al. 1997, Coveney et al. 2008). Migrating into the GR Leydig cells produce , required for the masculinization of the genitalia and maintenance of the . Sertoli cells secrete AMH, resulting in the degradation of the paramesonephric (Müllerian) duct. In the absence of , the develops as the , uterine tubules and upper portion of the . Female genitalia are also formed from the , folds and swellings. Post-natal oestradiol synthesis by the ovary is important for the final maturation of the oocytes and secondary sex characteristics. Other tissues of the body also show sex-dimorphic such as the brain and liver, under the influence of steroid hormones and other gonadal factors such as AMH (Yang et al. 2006, Wang et al. 2009, Conforto & Waxman 2012, Wittmann & McLennan 2013, Maekawa et al. 2014). the mammalian GR begins with increased proliferation of coelomic epithelial cells on the ventromedial surface of the mesonephroi. Each mesonephros also contains the mesonephric duct (also known as the Wolffian duct), a primordial urogenital tissue that will give rise to the male , and following male sex determination (Hannema & Hughes 2007, Shaw & Renfree 2014). In addition, the paramesonephric duct (also known as the Müllerian duct) is also present in the Figure 2 mesonephros, running in parallel to the mesonephric duct Schematic illustrations of the structures and components involved in early (Fig. 2A). This is the female equivalent to the mesonephric UGR development in mice. (A) (i) Side view of a E11.5 embryo indicating the duct, which will form the fallopian tubes, uterus and part location of the developing GR (orange bar). (ii) A ventral view of the abdominal region focuses on the mesonephros (white) and the gonadal of the following female sex determination (Acien ridge (grey) that develops on the ventromedial surface. The aorta is shown in 1992). Together, the mesonephros and GR are known as red. (iii) In transverse sections the mesonephros, containing mesonephric the urogenital ridge (UGR). and paramesonephric ducts, are often visible. The gonad appears as a bulge facing into the coelomic cavity. At this stage, the PGCs have migrated into the Proliferation of the coelomic epithelial cells on the GR, from the hindgut via the dorsal mesentery. (B) Starting at E9.5, the ventromedial surface of the mesonephros creates a dense, coelomic epithelium (yellow) on the ventromedial surface of the pseudostratified epithelial cell layer Gropp( & Ohno 1966, mesonephros begins to proliferate, forming a pseudostratified epithelial layer. The basement membrane becomes fragmented, allowing GR Pelliniemi 1975, Wartenberg et al. 1991). Alongside the progenitor cells that have undergone EMT to migrate inward. These cells proliferation of the coelomic epithelium, the underlying continue to proliferate, just behind the coelomic epithelium to form the basement membrane becomes fragmented, allowing bi-potential gonad. Between E10.0 and 11.5, the PGCs (green) also migrate into the GR. CE, coelomic epithelium; DM, dorsal mesentery; GC, germ cells; many epithelial cells to ingress dorsally, towards the GR, genital ridge; HG, hindgut; M, mesonephros; MD, mesonephric duct; MT, mesonephros (Fig. 2B) (Karl & Capel 1998, Kusaka et al. mesonephric tubule; PGC, primordial ; PMD, paramesonephric duct.

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R49 Endocrinology development just prior to gondal sex determination are primordial germ cluster of cells at the anterior region of cells (PGC), the precursors to the germ cells that become separates from the GR primordium and moves dorso- and oocytes later in life. PGCs arise near the yolk medially to form the adrenal anlage (Hatano et al. sac and travel, via the hindgut, to the GR as a result of 1996, Val & Swain 2010). From E11.5 to 12.5, neural chemotaxis (Doitsidou et al. 2002). crest cells invade the adrenal anlage and aggregate in the centre to form the medulla. Mesenchyme cells, hypothesized to be of a coelomic epithelial origin, Genital ridge formation in mice form a fibrous capsule around the composite adrenal Most of the work investigating mammalian gonad primordium by E14.5 (Xing et al. 2015). development has been performed on mice. It is assumed that events in the embryo, as well as other Development of the PGC in the Genital ridge Eutherian mammals, follow the same basic pattern, albeit with some differences in the timing and anatomy. As previously mentioned, PGCs originate from a separate The process of GR formation, starting with the location near the , away from the mesonephros proliferation of the coelomic epithelium, begins at about and GR (Saga 2008). Mammalian PGCs are specified via E9.5 in mice (Hu et al. 2013), which equates to about an inductive system of signalling molecules, particularly 5 weeks of gestation in humans (Jost 1972). The GR BMP4. In mice, PGCs are induced around E6.5 (equating itself is not morphologically evident until about E10.0– to approximately 2 weeks gestation in humans) from 10.5 where a clear distinction between the GR and the the proximal epiblast by BMP4 signalling (Lawson et al. mesonephros can be seen under light microscopy. From 1999). These cells subsequently move to and cluster at E10.5 to 11.5, further proliferation of the GR coelomic the base of the allantois/yolk sac wall, near the forming epithelium and mesonephros expands the overall size hindgut, which can be seen in mouse at about on the ventral side of the mesonephros (Fig. 2A). The E7.0 (week 3–4 human gestation) as a small population initiation, proliferation and expansion of the GR is of ~45 cells (Lawson et al. 1999). As development indistinguishable between XX and XY embryos, and proceeds, the hindgut folds and the PGCs migrate the size of the GR is determined by the length of the into the embryo proper (Hara et al. 2009, Harikae et al. embryonic trunk (Wainwright et al. 2014). At around 2013). By E9.5 the PGCs begin to migrate away from E11.5–12.0, the molecular events that determine gonad the hindgut towards the UGR and colonize the gonad fate occur, prompting the gonad to follow either a testis- between E10.0 and 11.0 (~5-week gestation in humans) or ovary-specific developmental trajectory from this (Fig. 2A; Witschi 1948, Molyneaux et al. 2001). During time-point. The first sign of of the this mass migration of PGCs, the hindgut descends into early differentiated gonad becomes evident in XY gonads the coelomic cavity and the last PGCs to migrate must at about E12.5, when testis cords can be seen (Hacker travel through the dorsal mesentery before entering the et al. 1995, Schmahl et al. 2000). gonads (Molyneaux et al. 2001). The GR is not only a progenitor tissue for the The PGCs undergo several rounds of to gonad, but also the , a critical endocrine achieve a population of about ~3000 cells by ~E11.5 (Tam tissue that synthesizes and secretes a variety of steroid & Snow 1981). Around this time, PGCs begin to undergo hormones to maintain body homeostasis and regulate a process known as licensing, undergoing a global the stress response (Yates et al. 2013, Walczak & change in gene expression, turning on required Hammer 2015). Consequently, several genes required for , concurrently switching off their for gonad development are also important for adrenal pluripotency genes over the course of the following days gland development (Luo et al. 1994, Gut et al. 2005, (Stebler et al. 2004, Gill et al. 2011, Rolland et al. 2011, Katoh-Fukui et al. 2005, Bandiera et al. 2013, Tevosian Seisenberger et al. 2012). Following this transition, the et al. 2015). The medulla and the cortex of the adult PGCs are referred to as gametogenesis-competent cells and have separate origins; the medulla is are poised to initiate either male or female differentiation, derived from neural crest cells, whereas the cortex is including meiosis, upon receiving cues from the somatic derived from cells located at the anterior-most region cells of either the forming testis or ovary and the nearby of the GR, indicating it has an intermediate mesonephric tissue (McLaren & Southee 1997, Adams & origin (Hatano et al. 1996). Beginning at E10.5, a small McLaren 2002, Gill et al. 2011).

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R50 Endocrinology development

Genes essential for initial gonad formation factors have mainly been characterized in mice, mutations in three of these genes (NR5A1, WT1 and GATA4) have also Mutational analysis using the mouse model, with some been found in patients with DSD, indicating a conserved additional evidence from human clinical cases, have role in reproductive development (Bashamboo et al. 2010b, brought to light a number of genes that are required to Kohler et al. 2011, Lourenco et al. 2011, Swartz et al. 2017). initiate the formation and proliferation of the GR, as well However, these genes are expressed and function in many as testis/ovary differentiation (Table 1). These include developing organ systems, meaning the loss of function Wilms’ tumour suppressor 1 (WT1) (Kreidberg et al. 1993, in both mice and humans produce a range of phenotypes Hammes et al. 2001), LIM gene 9 (LHX9) beyond the reproductive tract (Ingraham et al. 1994, (Birk et al. 2000), Nuclear Subfamily 5 Group Klamt et al. 1998, Hammes et al. 2001, Tevosian et al. A member 1 (NR5A1; also called 2015). Genes known to be required for GR development (SF1)) (Luo et al. 1994), empty spiracles homeobox gene often show altered expression in gene knockout lines, 2 (EMX2) (Miyamoto et al. 1997) and GATA-binding indicating a co-regulatory relationship exists between protein 4 (GATA4) (Hu et al. 2013). In mouse embryos, these critical factors (discussed further below). a homozygous null mutation in any one of these genes It is evident that many of these critical early factors causes gonadal agenesis. While the function of these five discussed below have multiple roles in reproductive

Table 1 Summary of genes essential for GR formation.

Gene Mouse UGR expression Mouse gonadal phenotype Human phenotype WT1 Expressed in the coelomic Degenerating gonad due to Frasier syndrome (Klamt et al. 1998) epithelium and mesonephros increased apoptosis (Kreidberg WAGR syndrome (Le Caignec et al. 2007) (Armstrong et al. 1993) et al. 1993, Hammes et al. 2001) Denys-Drash syndrome (Hastie 1992), Male WT1-KTS mice exhibit complete Meacham syndrome (Hammes et al. 2001) Phenotypes include streak gonads, Following sex determination, roles in ambiguous genitalia, hypospadias, 46, ovary and follicle development XY sex reversal (Suri et al. 2007) (Gao et al. 2014) Nr5a1 Initially expressed in the anterior Gonad regression due to increased Range of phenotypes including 46, XY region of the coelomic epithelium. apoptosis sex reversal, gonad dysgenesis, male Expression domain expands in an Loss of adrenal development, infertility, hypospadias, adrenal anterior–posterior direction (Hu obesity and pituitary abnormalities insufficiency, gonad dysgenesis, et al. 2013) XY KO mice show complete male-to- premature ovarian failure (reviewed in female sex reversal (Luo et al. 1994, Ferraz-de-Souza et al. 2011) Shinoda et al. 1995) 46, XX sex reversal, ovotestis (Bashamboo et al. 2016, Swartz et al. 2017) Emx2 Coelomic epithelium and cells that Absent gonads Endometriosis (Daftary & Taylor 2004) move into the underlying Reduced proliferation of the coelomic Endometrial cancer (Daftary & Taylor mesenchyme epithelium (Miyamoto et al. 1997, 2004) Mesonephric duct epithelia (Hu et al. Pellegrini et al. 1997) Incomplete Müllerian fusion (Liu et al. 2013) 2015) Lhx9 Coelomic epithelium and cells that Absent gonads None reported move into the underlying Reduced proliferation of the Limited evidence of involvement in mesenchyme (Birk et al. 2000, Hu coelomic epithelium (Birk et al. reproductive cancers (cervical cancer et al. 2013) 2000) promoter methylation (Bhat et al. 2017)) Gata4 Coelomic epithelium, expression Absent gonads Testicular abnormalities, ambiguous domain expands in an anterior– No expansion of the coelomic genitalia (Lourenco et al. 2011) posterior direction, precedes epithelium (Hu et al. 2013) Frequent mutation in ovarian cancers expression of Sf1 (Hu et al. 2013) Following sex determination, roles in (Cai et al. 2009) ovarian and testicular development (Efimenko et al. 2013) MAP3K1 Coelomic epithelium and gonad Minor testis abnormalities (Warr Ambiguous genitalia streak gonads mesenchyme (Warr et al. 2011) et al. 2011) (Pearlman et al. 2010, Granados et al. 2017)

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R51 Endocrinology development development, from formation of the bi-potential gonad of both sexes, to sex-specific roles following gonadal sex determination. Currently, we are lacking a detailed gene network integrating both the roles of these transcription factors, along with the signalling pathways that regulate them, to gain a broader picture of how these components together regulate cellular processes like differentiation and proliferation to control formation of the bi-potential gonad. Future -wide approaches to study gene interactions will help to better define the regulatory interactions between these proteins, and others, required for GR formation Figure 3 The Wt1 gene has multiple roles in gonad development and differentiation. Two isoforms of WT1, WT1(+KTS) and WT1( KTS) have Wilms tumour suppressor (Wt1) − differing roles in gonad development based on gene-knockout studies. Wt1 mRNA is expressed during gonad development Loss of WT1(+KTS) leads to reduced Sry gene expression and XY sex reversal, in contrast, deletion of WT1(−KTS) halts GR development for in the coelomic epithelium and mesonephros prior to both sexes (Hammes et al. 2001). However, if both splice forms of WT1 gonadal sex determination and in the progenitor support are present until E10.5, the gonad primordium still develops but with cells of both the developing testis and ovary (Armstrong altered cell fate, with most somatic cells now adopting a steroidogenic cell fate (Chen et al. 2017). et al. 1993). Wt1−/− mice have a recognizable gonad primordium at E11.0, but this then degenerates due to expression of Wt1 mRNA was similar to WT animals, due apoptosis of somatic cells (Kreidberg et al. 1993, Hammes to increased expression of the alternative splice form et al. 2001). Knockout mouse embryos, on a C57BL/6 compensating for the loss of the targeted splice form genetic background, do not survive to parturition due to (Hammes et al. 2001). Thus, this resulting shift in isoform embryonic lethality from heart defects, while knockout ratio (the ratio of −KTS/+KTS), leading to overexpression mice generated on other genetic backgrounds, such as of the alternative isoform, may also contribute to the Balb/c, survive until birth (Kreidberg et al. 1993, Herzer observed knockout phenotype. However, this has not et al. 1999). been investigated further in mice with transgenic The Wt1 gene encodes for a transcription mouse lines. In humans, an altered splice form factor that functions as either an activator or repressor ratio (increased WT1−KTS, reduced WT1+KTS isoform) of transcription; however, the structure and function does severely affect gonadal development resulting in of the WT1 protein varies depending on the cell type Frasier syndrome with streak gonads (Barbaux et al. 1997, and promoter used to transcribe the gene, RNA editing, Klamt et al. 1998). alternative usage of translational start sites and alternative More recently, Chen et al. (2017) showed that splicing (Bruening & Pelletier 1996, Scharnhorst et al. the conditional inactivation of Wt1 just prior to sex 1999, Dallosso et al. 2007). Of particular interest to determination at E10.5 allows gonadogenesis to proceed gonad development are two alternate splice forms of with reduced differentiation of Sertoli and granulosa cells the WT1 protein named WT1–KTS and WT1+KTS, due from somatic cell precursors. Thus, when Wt1 expression to either the inclusion or exclusion of three amino is lost from gonads at E13.5, most somatic cells develop acids located between the third and fourth zinc finger into steroidogenic cell types (Chen et al. 2017; Fig. 3). In domains (Hammes et al. 2001). Mice lacking expression contrast, in traditional knockout animals where Wt1 is of the Wt1(−KTS) isoform have gonads markedly reduced inactive throughout development, development of the GR in size and poorly differentiated (Hammes et al. 2001), is blocked (Kreidberg et al. 1993; (Fig. 3). Thus, the role of suggesting that WT1(−KTS) is the isoform required for the WT1 alters as gonad development progresses, from being proliferation and differentiation of GR cells. Knockout of required for initial cell proliferation and growth of the the Wt1(+KTS) splice transcript leads to complete male- GR, predominately through the WT1(−KTS) splice form, to-female sex reversal and reduced Sry gene expression, to controlling cell fate of somatic cells, and this is likely to yet, ovarian development proceeds normally (Hammes occur through direct regulation of Nr5a1 gene expression et al. 2001; Fig. 3). For both knockout lines, the overall (Fig. 4, described further in the following section).

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R52 Endocrinology development

not develop beyond the early GR stage, and as a result, XY mice show complete male-to-female sex reversal (Luo et al. 1994). Gonadal regression in Nr5a1−/− embryos is due to increased apoptosis of the somatic cells (Luo et al. 1994), also observed with Wt1 loss-of-function mice (Hammes et al. 2001). Recent single-cell RNA sequencing (scRNA-seq) of XY somatic cells, expressing Nr5a1 (Nr5a1-GFP labelled cells) prior to (E10.5) and following sex determination (E11.5–E16.5), confirmed the presence of a multipotent Nr5a1+ cell population in the GR that progressively forms both the supporting and steroidogenic cell lineages from E11.5 (Stevant et al. 2018). Regulation of Nr5a1 gene expression plays a critical role in gonad development, with all key transcription factors described to date in GR development function in regulating correct Nr5a1 expression (Fig. 4; Wilhelm & Englert 2002, Katoh-Fukui et al. 2005, Hu et al. 2013, Chen et al. 2017). LHX9 and WT1(−KTS) proteins both bind to Figure 4 the promoter region of Nr5a1 in vitro, and together they Summary of the molecular relationships during early gonad formation. (A) Summary of relationships between the core genes necessary for GR increase reporter gene expression in a Sertoli-like (TM4) development. Relationships are based on previous studies using cell line (Wilhelm & Englert 2002), a finding replicated in knockout lines where expression changes of genes were observed. Loss Leydig-like (TM3) and C2C12 (myoblast derived) cell lines of expression from knockout indicated a positive regulatory relationship and increased expression suggests a negative relationship. If direct (Val et al. 2007, Takasawa et al. 2014). However, in primary relationships are known, through studies such as ChIP-PCR and reporter Leydig cells WT1 overexpression repressed Nr5a1 gene assays (Wilhelm & Englert 2002, Katoh-Fukui et al. 2005, Franca et al. 2013, expression. Further experiments confirmed that WT1 Chen et al. 2017), they have been indicated by a solid line. (B) Recent studies suggest a complex regulatory relationship exist between WT1 and directly binds in vivo to the Nr5a1 promoter in Sertoli Nr5a1 (Chen et al. 2017). Nr5a1 expressing cells (Nr5a1+) in the GR cells obtained from 2-week-old mice (Chen et al. 2017). contribute to the Sertoli, interstitial and populations following It was suggested that these conflicting results were most sex determination. In the bi-potential GR, WT1 is required for Nr5a1 gene expression. In Sertoli cells WT1 binds directly to the Nr5a1 gene promoter likely due to the use of cell lines vs primary cells in these and appears to reduce its expression. In Leydig cells (no Wt1 expression), studies (Chen et al. 2017). The TM4 and TM3 cell lines Nr5a1 gene expression is significantly higher. Lhx9 is a candidate protein were derived from juvenile BALB/c mouse testis (Mather partner for WT1 (Wilhelm & Englert 2002), it is expressed in the GR, and later in interstitial cells. 1980) and both cell lines express a similar combination of cell type gene markers (Beverdam et al. 2003). It is also worth noting that Chen et al. (2017) used primary Steroidogenic factor 1 (Sf1), Leydig cells obtained from adult and juvenile mice on subfamily 5 group A member 1 (Nr5a1) a mixed background (C57Bl/6:129/SvEv), given that The Nr5a1 (Sf1) gene encodes a that strain background can strongly influence the resulting is expressed in the adrenal glands, coelomic epithelium, phenotypes (Herzer et al. 1999, Meeks et al. 2003, Brennan and anterior during & Capel 2004, Munger et al. 2009). WT1 also may have development (Luo et al. 1994, Morohashi & Omura differing roles in the regulation of Nr5a1 gene expression, 1996). During UGR development, Nr5a1 gene expression as the gonad develops into a testis, perhaps being required is limited to the gonad primordium, where it is required for an initial activation of Nr5a1, with protein partner for the proliferation and survival of progenitor somatic LHX9 in the early gonad and, following gonadal sex cells (Luo et al. 1994). After gonadal sex determination, determination, WT1 reduces Nr5a1 expression in those Nr5a1 gene expression is restricted to testis-specific NR5A1+ cells that are fated to become Sertoli cells. In this cells, namely Leydig and Sertoli cells (Luo et al. 1994). case, the presence or absence of certain protein partners, Nr5a1−/− mice show complete failure of adrenal and for instance LHX9, would impact on how WT1 regulates gonadal development, abnormalities of the pituitary and Nr5a1 gene expression. Previous studies have shown that hypothalamus and obesity (Luo et al. 1994, Shinoda et al. WT1 is converted from an activator to a repressor protein 1995). The gonads of Nr5a1-knockout mice embryos do by the protein partners such as BASP1 (McKay et al. 1999,

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R53 Endocrinology development

Carpenter et al. 2004). Given that NR5A1+ cells contribute mice embryos exhibit normal PGC migration into the UGR to both cell populations, it maybe that Nr5a1 expression region, but the initial thickening of the coelomic epithelium levels determine which fate, with Sertoli cells expressing is not prominent and the GR soon degenerates (Miyamoto significantly lower levels ofNr5a1 compared to Leydig et al. 1997). These mutants completely lack gonads, genital cells (Fig. 4B; Chen et al. 2017). tracts and kidneys (Miyamoto et al. 1997). The phenotype in Emx2−/− mice is a result of impaired cell migration from the coelomic epithelium through LIM homeobox 9 (Lhx9) the basement membrane, as well as increased apoptosis The Lhx9 gene encodes a transcription factor expressed (Kusaka et al. 2010). Interestingly, Nr5a1 gene expression in a variety of regions within the developing mouse is also significantly affected inEmx2 −/− embryos (Kusaka embryo, including the brain, heart, kidney, limb buds et al. 2010), suggesting that Emx2 acts upstream of Nr5a1 in and the coelomic epithelium (Retaux et al. 1999, Birk the developmental cascade, but how Emx2 regulates Nr5a1 et al. 2000, Failli et al. 2000, Molle et al. 2004, Oshima transcription, whether directly or indirectly remains unclear. et al. 2007, Smagulova et al. 2008, Tzchori et al. 2009, Yang & Wilson 2015). Lhx9 gene expression in the UGR GATA-binding protein 4 (Gata4) is first seen in the coelomic epithelium at E9.5 and later in the gonad primordium, until sexual differentiation GATA4 is a transcription factor that is essential for the where its expression becomes restricted to the interstitial/ development of multiple organs such as the heart, foregut, mesothelial regions of testes and the cortical regions of liver, ventral pancreas and the UGR (Kuo et al. 1997, (Birk et al. 2000). Lhx9−/− mutant mice exhibit a Molkentin et al. 1997, Viger et al. 1998, Hu et al. 2013). similar gonadal phenotype to that of Wt1(−KTS) −/− mice, Gata4 gene expression in the GR was originally linked to a whereby normal GR development and PGC migration is role in testis differentiation through activating transcription observed but discrete gonads fail to form and genetically of Sry together with WT1 (Tevosian et al. 2002). Later male mice show complete male-to-female sex reversal studies revealed that Gata4 is expressed prior to any other of their secondary sex characteristics (Birk et al. 2000). gonadal factor, initially in the anterior half of the coelomic The observed underdevelopment of the GR results from epithelium at E9.5 and expanding to the posterior region disrupted proliferation of the gonad primordium (Birk by E10.2 (Hu et al. 2013). Gata4-knockout mice show no et al. 2000), as opposed to increased apoptosis observed signs of gonadal initiation, with the coelomic epithelium in the Wt1−/− and Nr5a1−/− mice (Kreidberg et al. 1993, remaining as an undifferentiated monolayer, indicating Luo et al. 1994). In addition, male- and female-knockout that Gata4 is required for the initial thickening of the offspring are infertile, with atrophic uteri, and coelomic epithelial layer (Hu et al. 2013). In these knockout oviducts, no male accessory sex organs, increased follicle- mice, Lhx9 and Nr5a1 gene expression is lost, but Wt1 and stimulating hormone levels, and undetectable levels of Emx2 gene expression is unaffected (Hu et al. 2013). This testosterone and oestrogen (Birk et al. 2000). Interestingly, suggests that GATA4 acts not only upstream of the Lhx9 Lhx9−/− mice show no other phenotypic abnormalities gene, but also Nr5a1, possibly even directly regulating both outside of gonad agenesis and male-to-female sex reversal genes, as binding sites for GATA4 have been identified in the (Birk et al. 2000, Tzchori et al. 2009), despite expression proximal promoter regions of these two genes (Tremblay & in other tissues, such as the limbs, nervous system and Viger 2001, Smagulova et al. 2008, Hu et al. 2013). pancreas, during development. This may be a result of the functional redundancy of Lhx9 with its closely related Genes with minor but essential roles in paralogue, LIM homeobox gene, Lhx2 (Jurata & Gill 1998, Genital ridge development Birk et al. 2000, Tzchori et al. 2009). Other genes have been implicated in early GR development, although mice-knockout strains for these genes exhibit a Empty spiracles homeobox 2 (Emx2) less severe phenotype than that of the null phenotype of Emx2 encodes a transcription factor that is the mouse the key genes described earlier. While the loss of function homolog of the Drosophila head gap gene empty spiracles of the following genes does not cause termination of (ems). The Emx2 transcript is expressed in the dorsal gonad development, it does result in the underdevel­ telencephalon, mesonephros and coelomic epithelium opment of the gonads, often in combination with sex (Miyamoto et al. 1997, Yoshida et al. 1997). Emx2−/−-knockout reversal of the secondary sex characteristics.

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R54 Endocrinology development

Transcription factor 21 (Tcf21 or Pod1) downstream of Sry to promote expansion and repress steroidogenic cell lineages. TCF21, also known as epicardin, capsuling or transcription factor 21, belongs to the basic helix-loop-helix family Sine oculis-related homeobox 1/4 (Six1/Six4) of transcription factors. It is expressed by mesodermal cell types including heart proepicardial cells, kidney Six1 and Six4 genes belong to the mammalian homolog of and visceral smooth muscle as well as the endodermal the Drosophila sine oculis homeobox family of transcription gastrointestinal tract (Quaggin et al. 1998, 1999, Lu et al. factors, containing a distinctive SIX domain (required 2000, Miyagishi et al. 2000). Tcf21-knockout mice die for protein-protein interactions) and homeodomain in the perinatal period due to lung, kidney and cardiac (Kawakami et al. 2000). Six1 and Six4 genes are located defects, but also display gastric and splenic defects (Cui in the same genomic regions within about 100 kb of one et al. 2003, Funato et al. 2003, Plotkin & Mudunuri 2008). another, and have highly overlapping tissue expression In addition, Tcf21 XX and XY KO mice have irregular during mouse development (Boucher et al. 1996, Esteve shaped gonads, the urogenital tracts of both XX and XY & Bovolenta 1999, Ozaki et al. 2001, Fougerousse et al. mice are indistinguishable and XY pups had feminized 2002, Laclef et al. 2003). Six1/4 double KO mice show genitalia (Cui et al. 2004). Consistent with this phenotype, different phenotypes compared to either Six1 or Six4 Tcf21 gene expression is initially detected throughout the single KO mice, highlighting regions of redundant GR at the bi-potential stage and continues to be expressed function in the development of limbs, , in the gonads of both sexes, with expression slightly sensory neurons and kidney (Grifone et al. 2005, Konishi higher in the testes following sex determination (Tamura et al. 2006, Giordani et al. 2007, Kobayashi et al. 2007). et al. 2001). While the gonad does form in the Tcf21−/− The suggested functional redundancy between the two embryo, morphological defects such as a shorted length genes is further exhibited as both genes share a common were observed by E11, and vascular abnormalities were DNA-binding site, MEF3 (Kawakami et al. 2000, Kumar observed by E12.5 (Cui et al. 2004), suggesting a defect 2009). In particular, only Six1/4 double KO mice embryos early in gonadal development. Further studies focused on display smaller gonads and adrenal glands (Kobayashi expression analyses, these suggested a negative regulatory et al. 2007, Fujimoto et al. 2013). Out of the essential relationship exists between Tcf21 and Nr5a1, as Tcf21 KO GR genes listed previously, only Nr5a1 gene expression results in increased Nr5a1 expression and an expanded is significantly reduced, a finding corroborated through Leydig cell population (Miyagishi et al. 2000). Despite reporter assays showing that SIX1/4 is able to transactivate an increase in Leydig cell numbers, the XY KO genitalia Nr5a1 transcription (Fujimoto et al. 2013). Furthermore, it was feminized and the gonads fail to descend from an was also shown that SIX1/4 is able to activate Fog2 gene abdominal position, suggesting that testosterone levels expression and that interaction, together with GATA4, were low, either due to later apoptosis of the gonadal regulates Sry gene expression (Fujimoto et al. 2013). tissue or a not all steroidogenic enzymes were expressed Overall, this indicates that SIX1/4 is required necessary correctly (Cui et al. 2004). TCF21 also appears to repress for sufficient Nr5a1 gene expression in the early gonad Nr5a1 gene expression in the adrenal gland, with in vitro and, a loss of Six1/4 gene expression, reduces Nr5a1 gene studies revealing that TCF21 directly represses Nr5a1 gene expression, and this may be responsible for the Six1/4 expression, through binding at E-box sequences located KO undersized gonad phenotype. Nonetheless, there is in the Nr5a1 promoter (Franca et al. 2013, 2015). still sufficient Nr5a1 gene expression to form the gonad Interestingly, the Sertoli cells do differentiate in the primordium in Six1/4 KO embryos. Tcf21 KO line, but there was some evidence that this cell population was reduced in number (although it Chromobox homolog 2 (Cbx2) (mouse polycomb group was not quantified) Cui( et al. 2004). TCF21 can cause member, M33) a sex reversal-like phenotype in vitro using a rat primary ovarian system. Overexpression of Tcf21 CBX2 is a component of the polycomb group complex of causes these cells to express Sertoli-like gene markers, in regulatory proteins involved in the repression/silencing of a similar pattern to that observed with Sry overexpression genes. In mice, Cbx2−/−-knockout mice show XY gonadal (Bhandari et al. 2011). Additionally, the Tcf21 gene has male-to-female (testis-to-ovary) sex reversal, and XX proposed to be a direct target of SRY (Bhandari et al. animals have smaller or absent ovaries (Katoh-Fukui et al. 2011). Together, these results suggest that TCF1 acts 2005). Additionally, mutants show defects in adrenal and

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R55 Endocrinology development splenic development, which was shown to be a result of Pre-B-cell leukaemia homeobox 1 (Pbx1) a reduction in the expression of Nr5a1 (Katoh-Fukui et al. Pbx1 encodes a TALE (three amino acid loop extension) 2005). This has led to the suggestion that CBX2 acts as class homeodomain transcription factor that has been an upstream regulator of Nr5a1 gene expression. Cbx2 shown to be involved in a number of process during has also been identified as a factor contributing to the mammalian embryogenesis including, skeletal devel­ differentiation of the testis through indirect regulation of opment and patterning (Selleri et al. 2001), maintenance Sry (Katoh-Fukui et al. 2012). Genome-wide identification of haematopoiesis (DiMartino et al. 2001), pancreatic of CBX2 target genes in a human Sertoli-like cell line development (Kim et al. 2002) and kidney and adrenal suggests that CBX2 acts to stimulate male-specific genes, development (Schnabel et al. 2003). The GR of Pbx1-null while suppressing female-pathway genes (Eid et al. 2015). mice are also smaller due to decreased proliferation of In humans, a reported case study of a 46, XY child with the progenitor cells in the gonad primordium (Schnabel female internal and external genitalia and histologically et al. 2003). Further experiments found that this is due normal ovaries found a compound loss-of-function to the downregulation of Nr5a1 gene expression; PBX1 mutation in the coding region of CBX2 (Biason-Lauber also upregulates Nr5a1 during adrenal development et al. 2009). This study further lends support to the role of (Schnabel et al. 2003). The adrenal gland primordium is CBX2 in the trans-activation of Nr5a1 and its role in both initially part of the GR but then later buds off from the the GR and testis developmental pathways (Fig. 4A). very rostral end around E10.5 (Schnabel et al. 2003, Pitetti et al. 2013). Therefore, perturbed Pbx1 gene expression Insulin receptor, Insr and insulin-like growth factor prior to budding likely affects Nr5a1 expression in type 1 receptor (Igf1r) the rostral GR, if not the whole GR, causing reduced Nr5a1 expression and decreased proliferation of somatic Insulin and its related growth factors IGF1 and IGF2 regulate progenitor cells. a variety of physiological processes including metabolism, stimulation of cell proliferation, differentiation and survival (Efstratiadis 1998). Their function is mediated Odd-skipped related 1 (Odd1) by two membrane-associated tyrosine kinase receptors, the insulin receptor (INSR) and the IGF type 1 receptor Odd1 gene encodes a zinc finger transcription factor (IGF1R). The genes for Insr, Igf1r and insulin receptor- homologous to the Drosophila odd-skipped class of related receptor (Irr) have previously been shown to be transcription factors that are involved in embryonic necessary for the testis determination pathway, as mutant patterning and tissue morphogenesis (Wang et al. 2005). mice lacking all three genes show male-to-female sex Targeted gene knockout of Odd1 revealed that this gene reversal and decreased Sry and Sox9 gene expression (Nef functions in both heart and et al. 2003). Recently, it has been shown that Insr and Igf1r, development (Wang et al. 2005). Odd1−/− embryos but not Irr have roles in GR development, whereby mice have severe heart malformations, and completely lacking both Insr and Igf1r have reduced proliferation lack adrenal glands, kidneys and gonads, all of which of the GR prior to sex determination but also extensive derive from intermediate mesoderm (Wang et al. 2005). downregulation of hundreds of genes associated with Although gonad development was not the focus of adrenal, testicular and ovarian development (Pitetti et al. this study, they did show that in early development 2013). As a result, these mice embryos exhibit agenesis of the GR was hypoplastic (Wang et al. 2005), likely due the adrenal cortex, along with male-female sex reversal to increased apoptosis observed with the developing due to a delay in Sry gene upregulation. Interestingly, kidney. These hypoplastic GRs appeared to degenerate, ovarian differentiation is also delayed in these mice, as no visible gonad structures were observed by E15.5 leaving the GR in an undifferentiated state until about (Wang et al. 2005). Although no further investigation E16.5 when the ovarian programme is eventually initiated of Odd1 with regards to urogenital development (Pitetti et al. 2013). Among the genes downregulated are has been done since Wang et al. (2005), it is unclear Wt1, Lhx9 and Nr5a1, indicating that genes essential for if the GR phenotype was an effect of Odd1 acting GR development are under the influence of insulin/IGF either directly in the GR or indirectly, potentially signalling (Pitetti et al. 2013), but only partially dependent acting via downregulation of Wt1 gene expression, on this signalling pathway, as GR development is only which also has an apoptotic phenotype when deleted hindered by decreased progenitor cell numbers. (Hammes et al. 2001).

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R56 Endocrinology development

Genital ridge formation in humans

Much of what is known of human gonadal development is based on early work and case studies of individuals with abnormal characteristics stemming from improper sexual development as a result of disruption of genes involved in this process. Recently, as sequencing Figure 5 technologies have become more affordable and require Histology sections through human embryos 36–44 days ( 14–18). At ~36 days the coelomic epithelium begins to thicken, and by less material, more studies are using human-derived 39 days a ridge of tissue is forming, facing into the gut cavity. Just prior to samples to study human sex development at the molecular sex determination (~42 days), the mesenchyme has proliferated to form a level, mostly through studies of cases of disorders of gonad region that is now easily distinguished from the neighbouring mesonephros. Section images were obtained from the Virtual Human sexual development (DSD) (O’Shaughnessy et al. 2007, Embryo resource (https://www.prenatalorigins.org/virtual-human- Ostrer et al. 2007, Houmard et al. 2009, Del Valle et al. embryo/). CE, coelomic epithelium; DM, dorsal mesentery; M, 2017, Li et al. 2017). These are congenital disorders that mesonephros; MT, mesonephric tubule; MV, mesonephric vesicle; PMD, paramesonephric duct. Scale bar = 100 µm. arise as a consequence of atypical chromosomal, gonadal or anatomical sexual development (Hughes et al. 2006), development typically results in a range of phenotypes from including some of the aforementioned genes involved complete gonad dysgenesis to adult infertility. Mutations in GR development. The term DSD replaced old and in some of these early gonad development genes are also ambiguous terms such as , hermaphroditism and responsible for some cases of human idiopathic infertility pseudo-hermaphroditism previously used to describe in cases with apparently normal gonad development. such disorders in humans (Dreger et al. 2005, Lees & Like DSDs, infertility is a condition associated with severe Tuch 2006). DSD phenotypes include a broad range of emotional and mental stress, particularly in societies where conditions such as failure of gonad formation, mixed there is a social emphasis placed upon the ideal of having gonadal tissue (male- and female-specific cell types within biological children (Ashraf et al. 2014). The mechanisms the tissue), ambiguous genitalia and failure of secondary of infertility in males and are varied in both sexual characteristics to develop normally (Cools et al. origin and functional impact and often these conditions 2006). There is limited data on the incidence of DSDs, are thought to have an underlying genetic component but it is estimated that the overall worldwide incidence (Zorrilla & Yatsenko 2013). Only 6–18% of infertility cases of DSDs is 1 in 5,500 (Damiani 2007, Houk & Lee 2008). have identifiable genetic causes (mainly sex- In humans, the gonad develops from the coelomic abnormalities) indicating that for many cases of infertility epithelium (Fig. 5), and the first signs of sex differentiation (and subfertility), like DSD, the underlying genetic factors are observed between 6 and 7 weeks with the formation of are yet to be elucidated. testicular cords (Gruenwald 1942, Wyndham 1943). Germ Genes with mutations commonly identified in DSD cells are first detected in the hindgut dorsal mesentery at studies include receptor and synthesis genes 4 weeks (Carnegie Stage 12 (CS12)) and migrate into the ( (AR), CYP17A1, SRD5A2), NR5A1(SF1), GR by 5 weeks (CS16) (Mckay et al. 1953). Gondal sex WT1, GATA4, SRY, DAX1 and CHD7 (Kremen et al. 2017). determination occurs around 41–42 days in human embryos, Those patients with DSD and a complete gonad dysgenesis signified by the upregulation ofSRY gene expression in XY phenotype carry mutations in SRY, MAP3K1, DHH and embryos ((Hanley et al. 2000, Del Valle et al. 2017, Mamsen NR5A1 genes (Ono & Harley 2013). While these genes et al. 2017). Genes required for steroidogenesis and secreted are also critical for mouse gonadal development, largely factors such as AMH are essential for the sex-specific supporting the use of the mouse as a model for human development of genitalia and the reproductive tract, with gonad development, there are several exceptions. For their expression commencing between 54 and 57 dpc example, sequencing screens have found gain-of-function (CS23). Genes essential for GR development in mice are mutations in the mitogen-activated protein kinase kinase also expressed at similar levels in both XX and XY gonads kinase 1 (MAP3K1) gene, including patients with streak (LHX9, EMX2, WT1 and GATA4), further supporting a role gonads and female genitalia (Pearlman et al. 2010, Loke for these factors in early human gonad development (Del et al. 2014, Baxter et al. 2015, Eggers et al. 2016, Granados Valle et al. 2017, Mamsen et al. 2017). et al. 2017), suggesting a requirement for MAP3K1 Despite their apparent early essential roles in gonad function in gonad development for both sexes. However, development, the loss of function of genes required for sex although the mouse orthologue, Map3k1, is expressed in

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R57 Endocrinology development the E11.5 gonad mouse, knockout mice have only minor The use of single-cell RNA-seq (scRNA-seq) has rapidly testicular abnormalities, indicating a minimal role in XY furthered our understanding of developmental events, gonadal development (Warr et al. 2011). This stresses the particularly within heterogeneous cell populations importance of additionally developing mouse models that (Shapiro et al. 2013). Given that the GR consists of a broad replicate human gene mutations to study phenotype, as variety of cellular precursors for endothelial, steroidogenic often human gene mutations do not result in a complete and supporting cell lineages, along with maturing germ loss of gene/protein function but rather gain of function. cells, it is especially suitable for scRNA-seq analysis. Li Mutations in NR5A1 were identified in 4% of patients et al. undertook scRNA-seq analysis of isolated single examined with unexplained male infertility (Bashamboo foetal germ cells and their surrounding ‘niche’ somatic et al. 2010a, Ropke et al. 2013) and in female patients cells from 4- to 26-week-old human embryos (Li et al. within premature ovarian failure (Lourenco et al. 2009). 2017). Based on gene expression profiles from this study, Novel NR5A1 mutation I (R92W) leads to 46, XX ovotestis somatic cells in the early gonad can be divided into four (SRY negative) (Igarashi et al. 2017, Baetens et al. 2017b). groups within each sex. XX gonadal cells are comprised of The types of NR5A1 gene mutations and phenotypes endothelial cells, and three types of maturing granulosa associated with these disorders are reviewed in (Ferraz-de- cells (early-(10 weeks), mid-(10–20 weeks) and late- Souza et al. 2011). WT1 mutations have also been found granulosa (20–26 weeks)) (Li et al. 2017). In XY gonads, in male and female cases of infertility, with ‘normal’ somatic cells group into Sertoli cells, Leydig cell precursors, gonadal development (Seabra et al. 2015, Nathan et al. differentiated Leydig cells and endothelial cells (Li et al. 2017). Humans are not the only species to present with 2017). Systematic examination of the expression profiles variable phenotypes; in mice, the genetic background of each gonadal cell population during development will or strain strongly influences the adult phenotype of not only improve our knowledge of the in vivo mechanisms many genes linked to gonadal development. In one such of cell differentiation but also the conditions required to example, gene knockout of Gadd45g (growth arrest and induce correct cell differentiation in vitro. DNA damage-inducible protein 45 g) on a mixed genetic Organoid systems are becoming popular in vitro models background (129/C57BL/6), 20% of the XY homozygous of organ development (Fatehullah et al. 2016). Isolated mice developed as infertile males, whereas on a C57BL/6 preparations of human somatic and germ cells can self- (B6) background, 100% of XY mice were sex reversed organize into a testicular-like organoid using an artificial (Johnen et al. 2013). Genetic background also influences scaffold to aid 3D organization (Baert et al. 2017). These the phenotype of Nr5a1-, Fgf9- and Wt1-null mice (Meeks studies make use of cells that have already undergone et al. 2003, Brennan & Capel 2004). The B6 mouse strain sex-specific differentiation, as the cells are isolated from is more likely to result in a male-to-female sex reversal post-natal tissues (Baert et al. 2017). Recently, Sepponen phenotype than the 129S1/SvImJ (129S1) strain, and et al., reported using human embryonic stem cells (hESCs) differences in gonadal gene expression between these culture conditions to sequentially induce the primitive strains are observed even prior to sex determination at streak, followed by intermediate mesoderm and finally E11.5 (Colvin et al. 2001, Munger et al. 2009). Therefore, bi-potential-like gonadal cells expressing genes such as even though studies have identified genes critical to the LHX9, EMX2, WT1 and GATA4 (Sepponen et al. 2017). early steps of gonad development, a loss-of-function This study found that timing and levels of BMPs (bone mutation does not necessarily lead to complete gonadal morphogenetic protein), WNT/β-catenin and Activin-A dysgenesis. In at least some cases, genetic background and signalling ligands are essential to promote differentiation possibly environmental factors determine the phenotypic of gonadal cell precursors, over other mesodermal cell consequences of loss-of-function mutations in these genes. types (Sepponen et al. 2017). These studies, along with new sequencing resources, lay the groundwork for future research for not only modelling the early events of human Future directions: unravelling the molecular gonad development, but to also examine the functional pathways of early gonad development consequences of gene mutations identified in DSD The advent and affordability of high-throughput sequenc­ patients using cultured cells engineered with the same ing technologies and new methods of replicating organ genetic mutation. development in vitro, together will be valuable research Several groups have taken a comprehensive targeted tools in propelling forward both genetic and cellular screening approach in order to identify genetic factors in into early GR development. DSD patients, and this in turn may lead to the identification

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R58 Endocrinology development of novel genes required for GR development. Exome changes associated with DSD (reviewed in (Baetens et al. sequencing and targeted gene sequencing identified the 2017a). As putative regulatory elements are difficult to genetic cause (classed as a functional gene mutation) in predict, few have been mapped for human genes associated 28–38% of cases examined (Baxter et al. 2015, Dong et al. with early gonadal development. Recent genome-wide 2016, Fan et al. 2017, Kim et al. 2017). Deep sequencing of studies, including those mapping the state in 64 known and 967 candidate genes improved the genetic specific cell lineages (Mikkelsen et al. 2007), will improve diagnosis rate to 43% for one patient cohort (Eggers et al. our ability to predict if DNA changes associated with DSD 2016). Thus, despite advances in sequencing technologies, lie within gene regulatory regions. over 50% of DSD cases remain without a genetic diagnosis. Epigenetic mechanisms such as DNA methylation While targeted sequencing and exome sequencing and modifications have been implicated in the studies have focused on the protein coding regions of the regulation of Sry gene in mice and dogs (Nishino et al. genome, epigenetic processes, gene regulatory elements 2011, Jeong et al. 2016, Kuroki et al. 2017). Loss of function and non-coding RNAs (ncRNAs) are just as important of the JMJD1A gene, which encodes a H3K9 demethylase for correct embryonic development. Errors in any of enzyme, results in XY sex reversal (Kuroki et al. 2017). these gene regulatory mechanisms may underlie many Some cases of XY DSD with complete sex reversal in dogs cases of idiopathic DSD that lack mutations in protein- are thought to be due to persistent DNA hypermethylation coding genes. Studies in vertebrates have largely focused of the Sry gene (Jeong et al. 2016). Therefore, it is likely on identifying sex-dimorphic expression of small ncRNAs that some cases of human DSD may be the result of called miRNAs following sex determination (Bannister et al. mutations to epigenetic regulatory factors. Regulation of 2009, Real et al. 2013, Wainwright et al. 2013, Presslauer gene expression through epigenetic mechanisms is also et al. 2017). There is limited information regarding how especially sensitive to environmental influences and this these miRNAs function in sex determination, if their impacts on many developmental programmes including function is essential, if they act to finely adjust gene sex determination (Feil & Fraga 2012). DNA methylation expression levels during gonad development and whether levels, determined by environmental factors, are vital these, and others, have earlier roles in the formation of to many naturally occurring forms of sex reversal and the bi-potential gonad. Currently no miRNAs have been environmental sex determination in animals (Capel linked to human gonad development. Long ncRNAs are 2017). While difficult to study with respect to human DSD, another important class of ncRNAs, which function to it is possible that DNA methylation may play a role in regulate gene expression (reviewed in Moran et al. 2012). balancing one sex developmental trajectory over another, With respect to sex development, these have been most and thus, errors in this may lead to gonad dysgenesis or thoroughly investigated for their role in X-chromosome sex reversal in humans. dosage compensation (Cerase et al. 2015). It remains to be determined if autosomally encoded long ncRNAs Summary contribute to mammalian sex determination and gonadal development but given their important roles in the The complexity of reproductive development is reflected development of other organ systems, it is likely they have in the difficulty in assigning a genetic diagnosis in a role in some aspects of gonadal development. most cases of human DSD. While we know the genetic Genome regulatory elements have been difficult to aetiology of a small number of DSDs, up to as many as identify as most lie within the non-coding regions of the 75% of individuals with a DSD will remain without genome and can act over long distances to influence gene a genetic diagnosis (Arboleda et al. 2014). Even with expression via chromatin folding. Mutations in regulatory whole genome sequencing, it is often difficult to identify elements range from single nucleotide sequence variants functional variants and causal mutations, rendering that prevent or reduce binding of a transcription factor many sequencing approaches somewhat ineffective to their DNA target sequence, to larger deletions, (Fan et al. 2017). Gene expression levels, epigenetic duplications and translocations resulting in structural modifiers and genetic background, along with the type changes that alter chromatin confirmation and regulatory of mutation and its functional consequence can all interactions between enhancers and their target genes. influence the resulting phenotype for both humans and Loss-of-function mutations in regulatory elements mice. The future development of new technologies and located near the SOX9 gene, a gene important for male improvement of existing ones will provide us with a much sex determination, are the best-characterized regulatory better understanding of the processes underlying normal

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R59 Endocrinology development gonadal development in both human and mouse models, Barbaux S, Niaudet P, Gubler MC, Grunfeld JP, Jaubert F, Kuttenn F, which in turn will lead to improved diagnosis in cases of Fekete CN, Souleyreau-Therville N, Thibaud E, Fellous M, et al. 1997 Donor splice-site mutations in WT1 are responsible for Frasier DSD and infertility. syndrome. Nature Genetics 17 467–470. (https://doi.org/10.1038/ ng1297-467) Bashamboo A, Ferraz-de-Souza B, Lourenco D, Lin L, Sebire NJ, Montjean D, Bignon-Topalovic J, Mandelbaum J, Siffroi JP, Christin- Declaration of interest Maitre S, et al. 2010a Human male infertility associated with The authors declare that there is no conflict of interest that could be mutations in NR5A1 encoding steroidogenic factor 1. American perceived as prejudicing the impartiality of this review. Journal of Human Genetics 87 505–512. (https://doi.org/10.1016/j. ajhg.2010.09.009) Bashamboo A, Lourenco D, Rybczynska M, Nihoul-Fekete C, Brauner R & McElreavey K 2010b Familial case of 46, XY disorder of sex Funding development DSD and congenital heart defects (CHD) associated This work did not receive any specific grant from any funding agency in the with a mutation in GATA4. Endocrine Reviews 31 S1667. public, commercial, or not-for-profit sector. Bashamboo A, Donohoue PA, Vilain E, Rojo S, Calvel P, Seneviratne SN & Mulukutla SN 2016 A recurrent p. Arg92Trp variant in steroidogenic factor-1 (NR5A1) can act as a molecular switch in human sex development. Human Molecular Genetics 25 3446–3453. Acknowledgements (https://doi.org/10.1093/hmg/ddw390) The authors would like to thank James Smith, Kathy Sircombe, Rebecca Baxter RM, Arboleda VA, Lee H, Barseghyan H, Adam MP, Fechner PY, Clarke and Susie Szakats for their feedback on earlier versions of this Bargman R, Keegan C, Travers S, Schelley S, et al. 2015 Exome manuscript. Yisheng Yang was supported by a University of Otago PhD sequencing for the diagnosis of 46,XY disorders of sex development. scholarship award. Our work was funded by a University of Otago Research Journal of Clinical Endocrinology and Metabolism 100 E333–E344. Grant to Megan Wilson. (https://doi.org/10.1210/jc.2014-2605) Beverdam A, Wilhelm D & Koopman P 2003 Molecular characterization of three gonad cell lines. Cytogenetic and Genome Research 101 References 242–249. (https://doi.org/10.1159/000074344) Bhandari RK, Sadler-Riggleman I, Clement TM & Skinner MK 2011 Basic Acien P 1992 Embryological observations on the female genital tract. helix-loop-helix transcription factor TCF21 is a downstream target of Human Reproduction 7 437–445. (https://doi.org/10.1093/ the male sex determining gene SRY. PLoS ONE 6 e19935. (https://doi. oxfordjournals.humrep.a137666) org/10.1371/journal.pone.0019935) Adams IR & McLaren A 2002 Sexually dimorphic development of mouse Bhat S, Kabekkodu SP, Varghese VK, Chakrabarty S, Mallya SP, Rotti HS, primordial germ cells: switching from to spermatogenesis. Pandey D, Kushtagi P & Satyamoorthy K 2017 Aberrant gene-specific Development 129 1155–1164. DNA methylation signature analysis in cervical cancer. Tumor Biology Arboleda VA, Sandberg DE & Vilain E 2014 DSDs: genetics, underlying 39 1010428317694573. (https://doi.org/10.1177/1010428317694573) pathologies and psychosexual differentiation. Nature Reviews Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C & Schoenle EJ 2009 Endocrinology 10 603–615. (https://doi.org/10.1038/nrendo.2014.130) Ovaries and female phenotype in a girl with 46,XY karyotype and Armstrong JF, Pritchard-Jones K, Bickmore WA, Hastie ND & Bard JB mutations in the CBX2 gene. American Journal of Human Genetics 84 1993 The expression of the Wilms’ tumour gene, WT1, in the 658–663. (https://doi.org/10.1016/j.ajhg.2009.03.016) developing mammalian embryo. Mechanisms of Development 40 Birk OS, Casiano DE, Wassif CA, Cogliati T, Zhao L, Zhao Y, Grinberg A, 85–97. (https://doi.org/10.1016/0925-4773(93)90090-K) Huang S, Kreidberg JA, Parker KL, et al. 2000 The LIM homeobox Ashraf DM, Ali D & Azadeh DM 2014 Effect of infertility on the quality gene Lhx9 is essential for mouse gonad formation. Nature 403 of life, a cross-sectional study. Journal of Clinical and Diagnostic 909–913. (https://doi.org/10.1038/35002622) Research 8 OC13–OC15. Boucher CA, Carey N, Edwards YH, Siciliano MJ & Johnson KJ 1996 Baert Y, De Kock J, Alves-Lopes JP, Soder O, Stukenborg JB & Goossens E Cloning of the human SIX1 gene and its assignment to chromosome 2017 Primary human testicular cells self-organize into organoids 14. Genomics 33 140–142. (https://doi.org/10.1006/geno.1996.0172) with testicular properties. Reports 8 30–38. (https://doi. Brennan J & Capel B 2004 One tissue, two fates: molecular genetic org/10.1016/j.stemcr.2016.11.012) events that underlie testis versus ovary development. Nature Reviews Baetens D, Mendonca BB, Verdin H, Cools M & De Baere E 2017a Non- Genetics 5 509–521. (https://doi.org/10.1038/nrg1381) coding variation in disorders of sex development. Clinical Genetics Bruening W & Pelletier J 1996 A non-AUG translational initiation event 91 163–172. (https://doi.org/10.1111/cge.12911) generates novel WT1 isoforms. Journal of Biological Chemistry 271 Baetens D, Stoop H, Peelman F, Todeschini AL, Rosseel T, Coppieters F, 8646–8654. (https://doi.org/10.1074/jbc.271.15.8646) Veitia RA, Looijenga LH, De Baere E & Cools M 2017b NR5A1 is a Cai KQ, Caslini C, Capo-Chichi CD, Slater C, Smith ER, Wu H & Xu XX novel disease gene for 46,XX testicular and ovotesticular disorders of 2009 Loss of GATA4 and GATA6 expression specifies ovarian cancer sex development. Genetics in Medicine 19 367–376. (https://doi. histological subtypes and precedes neoplastic transformation of org/10.1038/gim.2016.118) ovarian surface epithelia. PLoS ONE 4 e6454. (https://10.1371/ Bandiera R, Vidal VP, Motamedi FJ, Clarkson M, Sahut-Barnola I, von journal.pone.0006454) Gise A, Pu WT, Hohenstein P, Martinez A & Schedl A 2013 WT1 Capel B 2017 Vertebrate sex determination: evolutionary plasticity of a maintains adrenal-gonadal primordium identity and marks a population fundamental switch. Nature Reviews Genetics 18 675–689. (https:// of AGP-like progenitors within the adrenal gland. Developmental Cell 27 doi.org/10.1038/nrg.2017.60) 5–18. (https://doi.org/10.1016/j.devcel.2013.09.003) Carpenter B, Hill KJ, Charalambous M, Wagner KJ, Lahiri D, James DI, Bannister SC, Tizard ML, Doran TJ, Sinclair AH & Smith CA 2009 Andersen JS, Schumacher V, Royer-Pokora B, Mann M, et al. 2004 Sexually dimorphic microRNA expression during chicken embryonic BASP1 is a transcriptional cosuppressor for the Wilms’ tumor gonadal development. Biology of Reproduction 81 165–176. (https:// suppressor protein WT1. Molecular and Cellular Biology 24 537–549. doi.org/10.1095/biolreprod.108.074005) (https://doi.org/10.1128/MCB.24.2.537-549.2004)

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R60 Endocrinology development

Cerase A, Pintacuda G, Tattermusch A & Avner P 2015 Xist localization Disorders of sex development: insights from targeted gene and function: new insights from multiple levels. Genome Biology 16 sequencing of a large international patient cohort. Genome Biology 166. (https://doi.org/10.1186/s13059-015-0733-y) 17 243. (https://doi.org/10.1186/s13059-016-1105-y) Chen M, Zhang L, Cui X, Lin X, Li Y, Wang Y, Wang Y, Qin Y, Chen D, Eid W, Opitz L & Biason-Lauber A 2015 Genome-wide identification of Han C, et al. 2017 Wt1 directs the lineage specification of sertoli and CBX2 targets: insights in the human sex development network. granulosa cells by repressing Sf1 expression. Development 144 44–53. Molecular Endocrinology 29 247–257. (https://doi.org/10.1210/ (https://doi.org/10.1242/dev.144105) me.2014-1339) Colvin JS, Green RP, Schmahl J, Capel B & Ornitz DM 2001 Male-to- Esteve P & Bovolenta P 1999 cSix4, a member of the six gene family of female sex reversal in mice lacking fibroblast growth factor 9. Cell transcription factors, is expressed during placode and somite 104 875–889. (https://doi.org/10.1016/S0092-8674(01)00284-7) development. Mechanisms of Development 85 161–165. (https://doi. Conforto TL & Waxman DJ 2012 Sex-specific mouse liver gene org/10.1016/S0925-4773(99)00079-9) expression: genome-wide analysis of developmental changes from Failli V, Rogard M, Mattei MG, Vernier P & Retaux S 2000 Lhx9 and pre-pubertal period to young adulthood. Biology of Sex Differences 3 Lhx9alpha LIM-homeodomain factors: genomic structure, expression 9. (https://doi.org/10.1186/2042-6410-3-9) patterns, chromosomal localization, and phylogenetic analysis. Cools M, Drop SL, Wolffenbuttel KP, Oosterhuis JW & Looijenga LH Genomics 64 307–317. (https://doi.org/10.1006/geno.2000.6123) 2006 Germ cell tumors in the intersex gonad: old paths, new Fan Y, Zhang X, Wang L, Wang R, Huang Z, Sun Y, Yao R, Huang X, Ye J, directions, moving frontiers. Endocrine Reviews 27 468–484. (https:// Han L, et al. 2017 Diagnostic application of targeted next-generation doi.org/10.1210/er.2006-0005) sequencing of 80 genes associated with disorders of sexual development. Coveney D, Cool J, Oliver T & Capel B 2008 Four-dimensional analysis of Scientific Reports 7 44536. (https://doi.org/10.1038/srep44536) vascularization during primary development of an organ, the gonad. Fatehullah A, Tan SH & Barker N 2016 Organoids as an in vitro model PNAS 105 7212–7217. (https://doi.org/10.1073/pnas.0707674105) of human development and disease. Nature Cell Biology 18 246–254. Cui S, Schwartz L & Quaggin SE 2003 Pod1 is required in stromal cells (https://doi.org/10.1038/ncb3312) for glomerulogenesis. Developmental Dynamics 226 512–522. (https:// Feil R & Fraga MF 2012 Epigenetics and the environment: emerging doi.org/10.1002/dvdy.10244) patterns and implications. Nature Reviews Genetics 13 97–109. Cui S, Ross A, Stallings N, Parker KL, Capel B & Quaggin SE 2004 Disrupted (https://doi.org/10.1038/nrg3142) gonadogenesis and male-to-female sex reversal in Pod1 knockout mice. Ferraz-de-Souza B, Lin L & Achermann JC 2011 Steroidogenic factor-1 Development 131 4095–4105. (https://doi.org/10.1242/dev.01266) (SF-1, NR5A1) and human disease. Molecular and Cellular Endocrinology Daftary GS & Taylor HS 2004 EMX2 gene expression in the female 336 198–205. (https://doi.org/10.1016/j.mce.2010.11.006) reproductive tract and aberrant expression in the of Fougerousse F, Durand M, Lopez S, Suel L, Demignon J, Thornton C, patients with endometriosis. Journal of Clinical Endocrinology and Ozaki H, Kawakami K, Barbet P, Beckmann JS, et al. 2002 Six and Eya Metabolism 89 2390–2396. (https://doi.org/10.1210/jc.2003-031389) expression during human somitogenesis and MyoD gene family Dallosso AR, Hancock AL, Malik S, Salpekar A, King-Underwood L, activation. Journal of Muscle Research and Cell Motility 23 255–264. Pritchard-Jones K, Peters J, Moorwood K, Ward A, Malik KT, et al. (https://doi.org/10.1023/A:1020990825644) 2007 Alternately spliced WT1 antisense transcripts interact with Franca MM, Ferraz-de-Souza B, Santos MG, Lerario AM, Fragoso MC, WT1 sense RNA and show epigenetic and splicing defects in cancer. Latronico AC, Kuick RD, Hammer GD & Lotfi CF 2013 POD-1 binding RNA 13 2287–2299. (https://doi.org/10.1261/rna.562907) to the E-box sequence inhibits SF-1 and StAR expression in human Damiani D 2007 Disorders of sexual development – still a big challenge! adrenocortical tumor cells. Molecular and Cellular Endocrinology 371 Journal of Pediatric Endocrinology and Metabolism 20 749–750. 140–147. (https://doi.org/10.1016/j.mce.2012.12.029) Del Valle I, Buonocore F, Duncan AJ, Lin L, Barenco M, Parnaik R, Franca MM, Abreu NP, Vrechi TA & Lotfi CF 2015 POD-1/Tcf21 Shah S, Hubank M, Gerrelli D & Achermann JC 2017 A genomic overexpression reduces endogenous SF-1 and StAR expression in rat atlas of human adrenal and gonad development. Wellcome Open adrenal cells. Brazilian Journal of Medical and Biological Research 48 Research 2 25. (https://doi.org/10.12688/wellcomeopenres.11253.1) 1087–1094. (https://doi.org/10.1590/1414-431X20154748) DiMartino JF, Selleri L, Traver D, Firpo MT, Rhee J, Warnke R, Fujimoto Y, Tanaka SS, Yamaguchi YL, Kobayashi H, Kuroki S, O’Gorman S, Weissman IL & Cleary ML 2001 The Hox cofactor and Tachibana M, Shinomura M, Kanai Y, Morohashi K, Kawakami K, proto-oncogene Pbx1 is required for maintenance of definitive et al. 2013 Homeoproteins Six1 and Six4 regulate male sex hematopoiesis in the fetal liver. Blood 98 618–626. (https://doi. determination and mouse gonadal development. Developmental Cell org/10.1182/blood.V98.3.618) 26 416–430. (https://doi.org/10.1016/j.devcel.2013.06.018) Doitsidou M, Reichman-Fried M, Stebler J, Koprunner M, Dorries J, Funato N, Ohyama K, Kuroda T & Nakamura M 2003 Basic helix-loop-helix Meyer D, Esguerra CV, Leung T & Raz E 2002 Guidance of primordial transcription factor epicardin/capsulin/Pod-1 suppresses differentiation germ cell migration by the chemokine SDF-1. Cell 111 647–659. by negative regulation of transcription. Journal of Biological Chemistry (https://doi.org/10.1016/S0092-8674(02)01135-2) 278 7486–7493. (https://doi.org/10.1074/jbc.M212248200) Dong Y, Yi Y, Yao H, Yang Z, Hu H, Liu J, Gao C, Zhang M, Zhou L, Gao F, Zhang J, Wang X, Yang J, Chen D, Huff V & Liu Y-x 2014 Wt1 Asan, et al. 2016 Targeted next-generation sequencing identification functions in development by regulating granulosa of mutations in patients with disorders of sex development. BMC cell differentiation. Human Molecular Genetics 23 333–341. (https:// Medical Genetics 17 23. (https://doi.org/10.1186/s12881-016-0286-2) doi.org/10.1093/hmg/ddt423) Dreger AD, Chase C, Sousa A, Gruppuso PA & Frader J 2005 Changing the Gill ME, Hu YC, Lin Y & Page DC 2011 Licensing of gametogenesis, nomenclature/taxonomy for intersex: a scientific and clinical rationale. dependent on RNA binding protein DAZL, as a gateway to sexual Journal of Pediatric Endocrinology and Metabolism 18 729–733. differentiation of fetal germ cells. PNAS 108 7443–7448. (https://doi. Efimenko E, Padua MB, Manuylov NL, Fox SC, Morse DA & Tevosian SG org/10.1073/pnas.1104501108) 2013 The transcription factor GATA4 is required for follicular Giordani J, Bajard L, Demignon J, Daubas P, Buckingham M & Maire P development and normal ovarian function. 2007 Six proteins regulate the activation of Myf5 expression in 381 144–158. (https://doi.org/10.1016/j.ydbio.2013.06.004) embryonic mouse limbs. PNAS 104 11310–11315. (https://doi. Efstratiadis A 1998 Genetics of mouse growth. International Journal of org/10.1073/pnas.0611299104) Developmental Biology 42 955–976. Granados A, Alaniz VI, Mohnach L, Barseghyan H, Vilain E, Ostrer H, Eggers S, Sadedin S, van den Bergen JA, Robevska G, Ohnesorg T, Quint EH, Chen M & Keegan CE 2017 MAP3K1-related gonadal Hewitt J, Lambeth L, Bouty A, Knarston IM, Tan TY, et al. 2016 dysgenesis: six new cases and review of the literature. American

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R61 Endocrinology development

Journal of Medical Genetics Part C: Seminars in Medical Genetics 175 Hughes IA, Houk C, Ahmed SF & Lee PA 2006 Consensus statement on 253–259. (https://doi.org/10.1002/ajmg.c.31559) management of intersex disorders. Journal of Pediatric Urology 2 Greenfield A 2015 Understanding sex determination in the mouse: 148–162. (https://doi.org/10.1016/j.jpurol.2006.03.004) genetics, epigenetics and the story of mutual antagonisms. Journal of Igarashi M, Takasawa K, Hakoda A, Kanno J, Takada S, Miyado M, Baba T, Genetics 94 585–590. (https://doi.org/10.1007/s12041-015-0565-2) Morohashi KI, Tajima T, Hata K, et al. 2017 Identical NR5A1 missense Grifone R, Demignon J, Houbron C, Souil E, Niro C, Seller MJ, Hamard G mutations in two unrelated 46,XX individuals with testicular tissues. & Maire P 2005 Six1 and Six4 homeoproteins are required for Pax3 Human Mutation 38 39–42. (https://doi.org/10.1002/humu.23116) and Mrf expression during in the mouse embryo. Ingraham HA, Lala DS, Ikeda Y, Luo X, Shen WH, Nachtigal MW, Abbud R, Development 132 2235–2249. (https://doi.org/10.1242/dev.01773) Nilson JH & Parker KL 1994 The nuclear receptor steroidogenic factor 1 Gropp A & Ohno S 1966 The presence of a common embryonic acts at multiple levels of the reproductive axis. Genes and Development 8 blastema for ovarian and testicular parenchymal (follicular, 2302–2312. (https://doi.org/10.1101/gad.8.19.2302) interstitial and tubular) cells in cattle Bos taurus. Zeitschrift für Ito M, Yokouchi K, Yoshida K, Kano K, Naito K, Miyazaki J & Tojo H Zellforschung und mikroskopische Anatomie 74 505–528. (https://doi. 2006 Investigation of the fate of Sry-expressing cells using an in vivo org/10.1007/BF00496841) Cre/loxP system. Development, Growth and Differentiation 48 41–47. Gruenwald P 1942 The development of the in the gonads of (https://doi.org/10.1111/j.1440-169X.2006.00844.x) and mammals. American Journal of Anatomy 70 359–397. Jeong YH, Lu H, Park CH, Li M, Luo H, Kim JJ, Liu S, Ko KH, Huang S, (https://doi.org/10.1002/aja.1000700303) Hwang IS, et al. 2016 Stochastic anomaly of methylome but Gut P, Huber K, Lohr J, Bruhl B, Oberle S, Treier M, Ernsberger U, persistent SRY hypermethylation in disorder of sex development in Kalcheim C & Unsicker K 2005 Lack of an adrenal cortex in Sf1 canine somatic cell nuclear transfer. Scientific Reports 6 31088. mutant mice is compatible with the generation and differentiation (https://doi.org/10.1038/srep31088) of chromaffin cells. Development 132 4611–4619. (https://doi. Johnen H, Gonzalez-Silva L, Carramolino L, Flores JM, Torres M & org/10.1242/dev.02052) Salvador JM 2013 Gadd45g is essential for primary sex Hacker A, Capel B, Goodfellow P & Lovell-Badge R 1995 Expression of determination, male fertility and testis development. PLoS ONE 8 Sry, the mouse sex determining gene. Development 121 1603–1614. e58751. (https://doi.org/10.1371/journal.pone.0058751) Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, Ziegler U, Jost A 1972 A new look at the mechanisms controlling sex differentiation Gubler MC & Schedl A 2001 Two splice variants of the Wilms’ in mammals. Johns Hopkins Medical Journal 130 38–53. tumor 1 gene have distinct functions during sex determination and Jurata LW & Gill GN 1998 Structure and function of LIM domains. nephron formation. Cell 106 319–329. (https://doi.org/10.1016/ Current Topics in Microbiology and Immunology 228 75–113. S0092-8674(01)00453-6) Karl J & Capel B 1995 Three-dimensional structure of the developing Hanley NA, Hagan DM, Clement-Jones M, Ball SG, Strachan T, Salas- mouse genital ridge. Philosophical Transactions of the Royal Society of Cortes L, McElreavey K, Lindsay S, Robson S, Bullen P, et al. 2000 London. Series B, Biological Sciences 350 235–242. (https://doi. SRY, SOX9, and DAX1 expression patterns during human sex org/10.1098/rstb.1995.0157) determination and gonadal development. Mechanisms of Development Karl J & Capel B 1998 Sertoli cells of the mouse testis originate from the 91 403–407. (https://doi.org/10.1016/S0925-4773(99)00307-X) coelomic epithelium. Developmental Biology 203 323–333. (https:// Hannema SE & Hughes IA 2007 Regulation of Wolffian duct doi.org/10.1006/dbio.1998.9068) development. Hormone Research 67 142–151. Katoh-Fukui Y, Owaki A, Toyama Y, Kusaka M, Shinohara Y, Maekawa M, Hara K, Kanai-Azuma M, Uemura M, Shitara H, Taya C, Yonekawa H, Toshimori K & Morohashi K 2005 Mouse Polycomb M33 is required Kawakami H, Tsunekawa N, Kurohmaru M & Kanai Y 2009 Evidence for splenic vascular and adrenal gland formation through regulating for crucial role of hindgut expansion in directing proper migration of Ad4BP/SF1 expression. Blood 106 1612–1620. (https://doi. primordial germ cells in mouse early embryogenesis. Developmental org/10.1182/blood-2004-08-3367) Biology 330 427–439. (https://doi.org/10.1016/j.ydbio.2009.04.012) Katoh-Fukui Y, Miyabayashi K, Komatsu T, Owaki A, Baba T, Shima Y, Harikae K, Miura K & Kanai Y 2013 Early gonadogenesis in mammals: Kidokoro T, Kanai Y, Schedl A, Wilhelm D, et al. 2012 Cbx2, a significance of long and narrow gonadal structure. Developmental polycomb group gene, is required for Sry gene expression in mice. Dynamics 242 330–338. (https://doi.org/10.1002/dvdy.23872) Endocrinology 153 913–924. (https://doi.org/10.1210/en.2011-1055) Hastie ND 1992 Dominant negative mutations in the Wilms tumour (WT1) Kawakami K, Sato S, Ozaki H & Ikeda K 2000 Six family genes--structure gene cause Denys-Drash syndrome – proof that a tumour-suppressor and function as transcription factors and their roles in development. gene plays a crucial role in normal genitourinary development. Human BioEssays 22 616–626. (https://doi.org/10.1002/1521- Molecular Genetics 1 293–295. (https://doi.org/10.1093/hmg/1.5.293) 1878(200007)22:7<616::AID-BIES4>3.0.CO;2-R) Hatano O, Takakusu A, Nomura M & Morohashi K 1996 Identical origin Kim SK, Selleri L, Lee JS, Zhang AY, Gu X, Jacobs Y & Cleary ML 2002 of adrenal cortex and gonad revealed by expression profiles of Pbx1 inactivation disrupts pancreas development and in Ipf1- Ad4BP/SF-1. Genes Cells 1 663–671. (https://doi. deficient mice promotes diabetes mellitus. Nature Genetics 30 org/10.1046/j.1365-2443.1996.00254.x) 430–435. (https://doi.org/10.1038/ng860) Herzer U, Crocoll A, Barton D, Howells N & Englert C 1999 The Wilms Kim JH, Kang E, Heo SH, Kim GH, Jang JH, Cho EH, Lee BH, Yoo HW & tumor suppressor gene is required for development of the Choi JH 2017 Diagnostic yield of targeted gene panel sequencing to spleen. Current Biology 9 837–840. (https://doi.org/10.1016/S0960- identify the genetic etiology of disorders of sex development. 9822(99)80369-8) Molecular and Cellular Endocrinology 444 19–25. (https://doi. Houk CP & Lee PA 2008 Consensus statement on terminology and org/10.1016/j.mce.2017.01.037) management: disorders of sex development. Sexual Development 2 Klamt B, Koziell A, Poulat F, Wieacker P, Scambler P, Berta P & Gessler M 172–180. (https://doi.org/10.1159/000152032) 1998 Frasier syndrome is caused by defective alternative splicing of Houmard B, Small C, Yang L, Naluai-Cecchini T, Cheng E, Hassold T & WT1 leading to an altered ratio of WT1 +/-KTS splice isoforms. Human Griswold M 2009 Global gene expression in the human fetal testis Molecular Genetics 7 709–714. (https://doi.org/10.1093/hmg/7.4.709) and ovary. Biology of Reproduction 81 438–443. (https://doi. Kobayashi H, Kawakami K, Asashima M & Nishinakamura R 2007 Six1 org/10.1095/biolreprod.108.075747) and Six4 are essential for Gdnf expression in the metanephric Hu YC, Okumura LM & Page DC 2013 Gata4 is required for formation mesenchyme and formation, while Six1 deficiency of the genital ridge in mice. PLoS Genetics 9 e1003629. (https://doi. alone causes mesonephric-tubule defects. Mechanisms of Development org/10.1371/journal.pgen.1003629) 124 290–303. (https://doi.org/10.1016/j.mod.2007.01.002)

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R62 Endocrinology development

Kohler B, Biebermann H, Friedsam V, Gellermann J, Maier RF, Pohl M, anomalies of human testicular development. PNAS 108 1597–1602. Wieacker P, Hiort O, Gruters A & Krude H 2011 Analysis of the (https://doi.org/10.1073/pnas.1010257108) Wilms’ tumor suppressor gene (WT1) in patients 46,XY disorders of Lu J, Chang P, Richardson JA, Gan L, Weiler H & Olson EN 2000 The sex development. Journal of Clinical Endocrinology and Metabolism 96 basic helix-loop-helix transcription factor capsulin controls spleen E1131–E1136. (https://doi.org/10.1210/jc.2010-2804) organogenesis. PNAS 97 9525–9530. (https://doi.org/10.1073/ Konishi Y, Ikeda K, Iwakura Y & Kawakami K 2006 Six1 and Six4 pnas.97.17.9525) promote survival of sensory neurons during early trigeminal Luo X, Ikeda Y & Parker KL 1994 A cell-specific nuclear receptor is gangliogenesis. Brain Research 1116 93–102. (https://doi. essential for adrenal and gonadal development and sexual org/10.1016/j.brainres.2006.07.103) differentiation. Cell 77 481–490. (https://doi.org/10.1016/0092- Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, Housman D & 8674(94)90211-9) Jaenisch R 1993 WT-1 is required for early . Cell Maekawa F, Tsukahara S, Kawashima T, Nohara K & Ohki-Hamazaki H 74 679–691. (https://doi.org/10.1016/0092-8674(93)90515-R) 2014 The mechanisms underlying sexual differentiation of behavior Kremen J, Chan YM & Swartz JM 2017 Recent findings on the genetics and physiology in mammals and birds: relative contributions of sex of disorders of sex development. Current Opinion in Urology 27 1–6. steroids and sex . Frontiers in Neuroscience 8 242. (https://doi.org/10.1097/MOU.0000000000000353) Mamsen LS, Ernst EH, Borup R, Larsen A, Olesen RH, Ernst E, Kumar JP 2009 The sine oculis homeobox (SIX) family of transcription Anderson RA, Kristensen SG & Andersen CY 2017 Temporal factors as regulators of development and disease. Cellular and Molecular expression pattern of genes during the period of sex differentiation Life Sciences 66 565–583. (https://doi.org/10.1007/s00018-008-8335-4) in human embryonic gonads. Scientific Reports 7 15961. (https://doi. Kuo CT, Morrisey EE, Anandappa R, Sigrist K, Lu MM, Parmacek MS, org/10.1038/s41598-017-15931-3) Soudais C & Leiden JM 1997 GATA4 transcription factor is required Martineau J, Nordqvist K, Tilmann C, Lovell-Badge R & Capel B 1997 for ventral morphogenesis and heart tube formation. Genes and Male-specific cell migration into the developing gonad. Current Development 11 1048–1060. (https://doi.org/10.1101/gad.11.8.1048) Biology 7 958–968. (https://doi.org/10.1016/S0960-9822(06)00415-5) Kuroki S, Okashita N, Baba S, Maeda R, Miyawaki S, Yano M, Mather JP 1980 Establishment and characterization of two distinct Yamaguchi M, Kitano S, Miyachi H, Itoh A, et al. 2017 Rescuing the mouse testicular epithelial cell lines. Biology of Reproduction 23 aberrant sex development of H3K9 demethylase Jmjd1a-deficient 243–252. (https://doi.org/10.1095/biolreprod23.1.243) mice by modulating H3K9 methylation balance. PLoS Genetics 13 Mckay DG, Hertig AT, Adams EC & Danziger S 1953 Histochemical e1007034. (https://doi.org/10.1371/journal.pgen.1007034) observations on the germ cells of human embryos. Anatomical Record Kusaka M, Katoh-Fukui Y, Ogawa H, Miyabayashi K, Baba T, Shima Y, 117 201–219. (https://doi.org/10.1002/ar.1091170206) Sugiyama N, Sugimoto Y, Okuno Y, Kodama R, et al. 2010 Abnormal McKay LM, Carpenter B & Roberts SG 1999 Regulation of the Wilms’ epithelial cell polarity and ectopic epidermal growth factor receptor tumour suppressor protein transcriptional activation domain. (EGFR) expression induced in Emx2 KO embryonic gonads. Oncogene 18 6546–6554. (https://doi.org/10.1038/sj.onc.1203046) Endocrinology 151 5893–5904. (https://doi.org/10.1210/en.2010-0915) McLaren A & Southee D 1997 Entry of mouse embryonic germ cells into Laclef C, Hamard G, Demignon J, Souil E, Houbron C & Maire P 2003 meiosis. Developmental Biology 187 107–113. (https://doi. Altered myogenesis in Six1-deficient mice. Development 130 org/10.1006/dbio.1997.8584) 2239–2252. (https://doi.org/10.1242/dev.00440) Meeks JJ, Weiss J & Jameson JL 2003 Dax1 is required for testis Lawson KA, Dunn NR, Roelen BA, Zeinstra LM, Davis AM, Wright CV, determination. Nature Genetics 34 32–33. (https://doi.org/10.1038/ Korving JP & Hogan BL 1999 Bmp4 is required for the generation of ng1141) primordial germ cells in the mouse embryo. Genes and Development Mikkelsen TS, Ku M, Jaffe DB, Issac B, Lieberman E, Giannoukos G, 13 424–436. (https://doi.org/10.1101/gad.13.4.424) Alvarez P, Brockman W, Kim TK, Koche RP, et al. 2007 Genome-wide Le Caignec C, Delnatte C, Vermeesch JR, Boceno M, Joubert M, maps of chromatin state in pluripotent and lineage-committed cells. Lavenan, F & Rival JM 2007 Complete sex reversal in a WAGR Nature 448 553–560. (https://doi.org/10.1038/nature06008) syndrome patient. American Journal of Medical Genetics Part A 143 Miyagishi M, Nakajima T & Fukamizu A 2000 Molecular characterization 2692–2695. (https://doi.org/10.1002/ajmg.a.31997) of mesoderm-restricted basic helix-loop-helix protein, POD-1/ Lees JG & Tuch BE 2006 Conversion of embryonic stem cells into Capsulin. International Journal of Molecular Medicine 5 27–31. pancreatic beta-cell surrogates guided by ontogeny. Regenerative Miyamoto N, Yoshida M, Kuratani S, Matsuo I & Aizawa S 1997 Defects Medicine, 1327–336. (https://doi.org/10.2217/17460751.1.3.327) of urogenital development in mice lacking Emx2. Development 124 Li L, Dong J, Yan L, Yong J, Liu X, Hu Y, Fan X, Wu X, Guo H, Wang X, 1653–1664. et al. 2017 Single-cell RNA-seq analysis maps development of human Molkentin JD, Lin Q, Duncan SA & Olson EN 1997 Requirement of the germline cells and gonadal niche interactions. Cell Stem Cell 20 858. transcription factor GATA4 for heart tube formation and ventral e854–873.e854. (https://doi.org/10.1016/j.stem.2017.05.009) morphogenesis. Genes and Development 11 1061–1072. (https://doi. Liu S, Gao X, Qin Y, Liu W, Huang T, Ma J & Chen ZJ 2015 Nonsense org/10.1101/gad.11.8.1061) mutation of EMX2 is potential causative for uterus didelphysis: first Molle B, Pere S, Failli V, Bach I & Retaux S 2004 Lhx9 and lhx9alpha: molecular explanation for isolated incomplete müllerian fusion. differential biochemical properties and effects on neuronal Fertility and Sterility 103 769–774. (https://doi.org/10.1016/j. differentiation. DNA Cell Biol 23 761–768. (https://doi.org/10.1089/ fertnstert.2014.11.03) dna.2004.23.761) Loke J, Pearlman A, Radi O, Zuffardi O, Giussani U, Pallotta R, Molyneaux KA, Stallock J, Schaible K & Wylie C 2001 Time-lapse Camerino G & Ostrer H 2014 Mutations in MAP3K1 tilt the balance analysis of living mouse germ cell migration. Developmental Biology from SOX9/FGF9 to WNT/beta-catenin signaling. Human Molecular 240 488–498. (https://doi.org/10.1006/dbio.2001.0436) Genetics 23 1073–1083. (https://doi.org/10.1093/hmg/ddt502) Moran VA, Perera RJ & Khalil AM 2012 Emerging functional and Lourenco D, Brauner R, Lin L, De Perdigo A, Weryha G, Muresan M, mechanistic paradigms of mammalian long non-coding RNAs. Nucleic Boudjenah R, Guerra-Junior G, Maciel-Guerra AT, Achermann JC, Acids Research 40 6391–6400. (https://doi.org/10.1093/nar/gks296) et al. 2009 Mutations in NR5A1 associated with ovarian Mork L, Maatouk DM, McMahon JA, Guo JJ, Zhang P, McMahon AP & insufficiency. New England Journal of Medicine 360 1200–1210. Capel B 2012 Temporal differences in specification in the (https://doi.org/10.1056/NEJMoa0806228) ovary reflect distinct follicle fates in mice. Biology of Reproduction 86 37. Lourenco D, Brauner R, Rybczynska M, Nihoul-Fekete C, McElreavey K Morohashi KI & Omura T 1996 Ad4BP/SF-1, a transcription factor & Bashamboo A 2011 Loss-of-function mutation in GATA4 causes essential for the transcription of steroidogenic cytochrome P450

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R63 Endocrinology development

genes and for the establishment of the reproductive function. FASEB Quaggin SE, Schwartz L, Cui S, Igarashi P, Deimling J, Post M & Journal 10 1569–1577. (https://doi.org/10.1096/fasebj.10.14.9002548) Rossant J 1999 The basic-helix-loop-helix protein pod1 is critically Munger SC, Aylor DL, Syed HA, Magwene PM, Threadgill DW & Capel B important for kidney and lung organogenesis. Development 126 2009 Elucidation of the transcription network governing 5771–5783. mammalian sex determination by exploiting strain-specific Real FM, Sekido R, Lupianez DG, Lovell-Badge R, Jimenez R & Burgos M susceptibility to sex reversal. Genes and Development 23 2521–2536. 2013 A microRNA (mmu-miR-124) prevents Sox9 expression in (https://doi.org/10.1101/gad.1835809) developing mouse ovarian cells. Biology of Reproduction 89 78. Nathan A, Reinhardt P, Kruspe D, Jorss T, Groth M, Nolte H, Retaux S, Rogard M, Bach I, Failli V & Besson MJ 1999 Lhx9: a novel Habenicht A, Herrmann J, Holschbach V, Toth B, et al. 2017 The LIM-homeodomain gene expressed in the developing forebrain. Wilms tumor protein Wt1 contributes to female fertility by Journal of Neuroscience 19 783–793. (https://doi.org/10.1523/ regulating oviductal proteostasis. Human Molecular Genetics 26 JNEUROSCI.19-02-00783.1999) 1694–1705. (https://doi.org/10.1093/hmg/ddx075) Rolland AD, Lehmann KP, Johnson KJ, Gaido KW & Koopman P 2011 Nef S, Verma-Kurvari S, Merenmies J, Vassalli JD, Efstratiadis A, Accili D Uncovering gene regulatory networks during mouse fetal germ cell & Parada LF 2003 Testis determination requires insulin receptor development. Biology of Reproduction 84 790–800. (https://doi. family function in mice. Nature 426 291–295. (https://doi. org/10.1095/biolreprod.110.088443) org/10.1038/nature02059) Ropke A, Tewes AC, Gromoll J, Kliesch S, Wieacker P & Tuttelmann F Nishino K, Hattori N, Sato S, Arai Y, Tanaka S, Nagy A & Shiota K 2011 2013 Comprehensive sequence analysis of the NR5A1 gene encoding Non-CpG methylation occurs in the regulatory region of the Sry steroidogenic factor 1 in a large group of infertile males. European gene. Journal of Reproduction and Development 57 586–593. (https:// Journal of Human Genetics 21 1012–1015. (https://doi.org/10.1038/ doi.org/10.1262/jrd.11-033A) ejhg.2012.290) Ono M & Harley VR 2013 Disorders of sex development: new genes, Saga Y 2008 Mouse germ cell development during embryogenesis. new concepts. Nature Reviews Endocrinology 9 79–91. (https://doi. Current Opinion in Genetics and Development 18 337–341. (https://doi. org/10.1038/nrendo.2012.235) org/10.1016/j.gde.2008.06.003) O’Shaughnessy PJ, Baker PJ, Monteiro A, Cassie S, Bhattacharya S & Scharnhorst V, Dekker P, van der Eb AJ & Jochemsen AG 1999 Internal Fowler PA 2007 Developmental changes in human fetal testicular cell translation initiation generates novel WT1 protein isoforms with numbers and messenger ribonucleic acid levels during the second distinct biological properties. Journal of Biological Chemistry 274 trimester. Journal of Clinical Endocrinology and Metabolism 92 4792–4801. 23456–23462. (https://doi.org/10.1074/jbc.274.33.23456) Oshima Y, Noguchi K & Nakamura M 2007 Expression of Lhx9 isoforms Schmahl J, Eicher EM, Washburn LL & Capel B 2000 Sry induces cell in the developing gonads of Rana rugosa. Zoological Science 24 proliferation in the mouse gonad. Development 127 65–73. 798–802. (https://doi.org/10.2108/zsj.24.798) Schnabel CA, Selleri L & Cleary ML 2003 Pbx1 is essential for adrenal Ostrer H, Huang HY, Masch RJ & Shapiro E 2007 A cellular study of development and urogenital differentiation. Genesis 37 123–130. human testis development. Sexual Development 1 286–292. (https:// (https://doi.org/10.1002/gene.10235) doi.org/10.1159/000108930) Seabra CM, Quental S, Lima AC, Carvalho F, Goncalves J, Fernandes S, Ozaki H, Watanabe Y, Takahashi K, Kitamura K, Tanaka A, Urase K, Pereira I, Silva J, Marques PI, Sousa M, et al. 2015 The mutational Momoi T, Sudo K, Sakagami J, Asano M, et al. 2001 Six4, a putative spectrum of WT1 in male infertility. Journal of Urology 193 myogenin gene regulator, is not essential for mouse embryonal 1709–1715. (https://doi.org/10.1016/j.juro.2014.11.004) development. Molecular and Cellular Biology 21 3343–3350. (https:// Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, Popp C, doi.org/10.1128/MCB.21.10.3343-3350.2001) Thienpont B, Dean W & Reik W 2012 The dynamics of genome- Pearlman A, Loke J, Le Caignec C, White S, Chin L, Friedman A, wide DNA methylation reprogramming in mouse primordial germ Warr N, Willan J, Brauer D, Farmer C, et al. 2010 Mutations in cells. Molecular Cell 48 849–862. (https://doi.org/10.1016/j. MAP3K1 cause 46,XY disorders of sex development and implicate a molcel.2012.11.001) common signal transduction pathway in human testis Selleri L, Depew MJ, Jacobs Y, Chanda SK, Tsang KY, Cheah KS, determination. American Journal of Human Genetics 87 898–904. Rubenstein JL, O’Gorman S & Cleary ML 2001 Requirement for Pbx1 (https://doi.org/10.1016/j.ajhg.2010.11.003) in skeletal patterning and programming chondrocyte proliferation Pellegrini M, Pantano S, Lucchini F, Fumi M & Forabosco A 1997 Emx2 and differentiation. Development 128 3543–3557. developmental expression in the primordia of the reproductive and Sepponen K, Lundin K, Knuus K, Vayrynen P, Raivio T, Tapanainen JS & excretory systems. Anatomy and Embryology 196 427–433. (https:// Tuuri T 2017 The role of sequential BMP signaling in directing doi.org/10.1007/s004290050110) human embryonic stem cells to bipotential gonadal cells. Journal of Pelliniemi LJ 1975 Ultrastructure of in male and female Clinical Endocrinology and Metabolism 102 4303–4314. pig embryos. Anatomy and Embryology 147 20–34. Shapiro E, Biezuner T & Linnarsson S 2013 Single-cell sequencing-based Pitetti JL, Calvel P, Romero Y, Conne B, Truong V, Papaioannou MD, technologies will revolutionize whole-organism science. Nature Schaad O, Docquier M, Herrera PL, Wilhelm D, et al. 2013 Insulin Reviews Genetics 14 618–630. (https://doi.org/10.1038/nrg3542) and IGF1 receptors are essential for XX and XY gonadal Shaw G & Renfree MB 2014 Wolffian duct development. Sexual differentiation and adrenal development in mice. PLoS Genetics 9 Development 8 273–280. (https://doi.org/10.1159/000363432) e1003160. (https://doi.org/10.1371/journal.pgen.1003160) Shinoda K, Lei H, Yoshii H, Nomura M, Nagano M, Shiba H, Sasaki H, Plotkin M & Mudunuri V 2008 Pod1 induces myofibroblast differentiation Osawa Y, Ninomiya Y, Niwa O, et al. 1995 Developmental defects of in mesenchymal progenitor cells from mouse kidney. Journal of Cellular the ventromedial hypothalamic nucleus and pituitary gonadotroph Biochemistry 103 675–690. (https://doi.org/10.1002/jcb.21441) in the Ftz-F1 disrupted mice. Developmental Dynamics 204 22–29. Presslauer C, Tilahun Bizuayehu T, Kopp M, Fernandes JM & Babiak I (https://doi.org/10.1002/aja.1002040104) 2017 Dynamics of miRNA transcriptome during gonadal Smagulova FO, Manuylov NL, Leach LL & Tevosian SG 2008 GATA4/ development of zebrafish. Scientific Reports 7 43850. (https://doi. FOG2 transcriptional complex regulates Lhx9 gene expression in org/10.1038/srep43850) murine heart development. BMC Developmental Biology 8 67. (https:// Quaggin SE, Vanden Heuvel GB & Igarashi P 1998 Pod-1, a mesoderm- doi.org/10.1186/1471-213X-8-67) specific basic-helix-loop-helix protein expressed in mesenchymal and Stebler J, Spieler D, Slanchev K, Molyneaux KA, Richter U, Cojocaru V, glomerular epithelial cells in the developing kidney. Mechanisms of Tarabykin V, Wylie C, Kessel M & Raz E 2004 Primordial germ cell Development 71 37–48. (https://doi.org/10.1016/S0925-4773(97)00201-3) migration in the chick and mouse embryo: the role of the

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access Journal of Molecular Y Yang et al. Mammalian early genital ridge 62:1 R64 Endocrinology development

chemokine SDF-1/CXCL12. Developmental Biology 272 351–361. et al. 2013 SOX9 regulates microRNA miR-202-5p/3p expression (https://doi.org/10.1016/j.ydbio.2004.05.009) during mouse testis differentiation. Biology of Reproduction 89 34. Stevant I, Neirijnck Y, Borel C, Escoffier J, Smith LB, Antonarakis SE, Wainwright EN, Svingen T, Ng ET, Wicking C & Koopman P 2014 Dermitzakis ET & Nef S 2018 Deciphering cell lineage specification Primary cilia function regulates the length of the embryonic trunk during male sex determination with single-cell RNA sequencing. Cell axis and urogenital field in mice. Developmental Biology 395 342–354. Reports 22 1589–1599. (https://doi.org/10.1016/j.celrep.2018.01.043) (https://doi.org/10.1016/j.ydbio.2014.08.037) Suri M, Kelehan P, O’Neill D, Vadeyar S, Grant J, Ahmed SF & FitzPatrick Walczak EM & Hammer GD 2015 Regulation of the adrenocortical stem D 2007 WT1 mutations in Meacham syndrome suggest a coelomic cell niche: implications for disease. Nature Reviews Endocrinology 11 mesothelial origin of the cardiac and diaphragmatic malformations. 14–28. (https://doi.org/10.1038/nrendo.2014.166) American Journal of Medical Genetics Part A 143 2312–2320. Wang Q, Lan Y, Cho ES, Maltby KM & Jiang R 2005 Odd-skipped related (https://doi.org/10.1002/ajmg.a.31924) 1 (Odd 1) is an essential regulator of heart and urogenital Swartz JM, Ciarlo R, Guo MH, Abrha A, Weaver B, Diamond DA, development. Developmental Biology 288 582–594. (https://doi. Chan YM & Hirschhorn JN 2017 A 46,XX ovotesticular disorder of org/10.1016/j.ydbio.2005.09.024) sex development likely caused by a steroidogenic factor-1 (NR5A1) Wang PY, Protheroe A, Clarkson AN, Imhoff F, Koishi K & McLennan IS variant. Hormone Research in Paediatrics 87 191–195. (https://doi. 2009 Mullerian inhibiting substance contributes to sex-linked biases org/10.1159/000452888) in the brain and behavior. PNAS 106 7203–7208. (https://doi. Takasawa K, Kashimada K, Pelosi E, Takagi M, Morio T, Asahara H, org/10.1073/pnas.0902253106) Schlessinger D, Mizutani S & Koopman P 2014 FOXL2 Warr N, Bogani D, Siggers P, Brixey R, Tateossian H, Dopplapudi A, transcriptionally represses Sf1 expression by antagonizing WT1 Wells S, Cheeseman M, Xia Y, Ostrer H, et al. 2011 Minor during ovarian development in mice. FASEB Journal 28 2020–2028. abnormalities of testis development in mice lacking the gene (https://doi.org/10.1096/fj.13-246108) encoding the MAPK signalling component, MAP3K1. PLoS ONE 6 Tam PP & Snow MH 1981 Proliferation and migration of primordial e19572. (https://doi.org/10.1371/journal.pone.0019572) germ cells during compensatory growth in mouse embryos. Journal Wartenberg H, Kinsky I, Viebahn C & Schmolke C 1991 Fine structural of Embryology and Experimental Morphology 64 133–147. characteristics of testicular cord formation in the developing rabbit Tamura M, Kanno Y, Chuma S, Saito T & Nakatsuji N 2001 Pod-1/ gonad. Journal of Electron Microscopy Technique 19 133–157. (https:// Capsulin shows a sex- and stage-dependent expression pattern in the doi.org/10.1002/jemt.1060190203) mouse gonad development and represses expression of Ad4BP/SF-1. Wilhelm D & Englert C 2002 The Wilms tumor suppressor WT1 Mechanisms of Development 102 135–144. (https://doi.org/10.1016/ regulates early gonad development by activation of Sf1. Genes and S0925-4773(01)00298-2) Development 16 1839–1851. (https://doi.org/10.1101/gad.220102) Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Eicher EM & Orkin SH Witschi E 1948 Migration of the germ cells of human embryos from the 2002 Gonadal differentiation, sex determination and normal Sry yolk sac to the primitive gonadal fold. Contributions to Embryology 32 expression in mice require direct interaction between transcription 57–80. partners GATA4 and FOG2. Development 129 4627–4634. Wittmann W & McLennan IS 2013 Anti-Mullerian hormone may Tevosian SG, Jimenez E, Hatch HM, Jiang T, Morse DA, Fox SC & regulate the number of calbindin-positive neurons in the sexually Padua MB 2015 adrenal development in mice requires GATA4 and dimorphic nucleus of the preoptic area of male mice. Biology of Sex GATA6 transcription factors. Endocrinology 156 2503–2517. (https:// Differences 4 18. (https://doi.org/10.1186/2042-6410-4-18) doi.org/10.1210/en.2014-1815) Wyndham NR 1943 A morphological study of testicular descent. Journal Tremblay JJ & Viger RS 2001 GATA factors differentially activate multiple of Anatomy 77 179-188.3. gonadal promoters through conserved GATA regulatory elements. Xing Y, Lerario AM, Rainey W & Hammer GD 2015 Development of Endocrinology 142 977–986. (https://doi.org/10.1210/endo.142.3.7995) adrenal cortex zonation. Endocrinology Metabolism Clinics of North Tzchori I, Day TF, Carolan PJ, Zhao Y, Wassif CA, Li L, Lewandoski M, America 44 243–274. (https://doi.org/10.1016/j.ecl.2015.02.001) Gorivodsky M, Love PE, Porter FD, et al. 2009 LIM homeobox Yang Y & Wilson MJ 2015 Lhx9 gene expression during early limb transcription factors integrate signaling events that control three- development in mice requires the FGF signalling pathway. Gene dimensional limb patterning and growth. Development 136 Expression Patterns 19 45–51. (https://doi.org/10.1016/j.gep.2015.07.002) 1375–1385. (https://doi.org/10.1242/dev.026476) Yang X, Schadt EE, Wang S, Wang H, Arnold AP, Ingram-Drake L, Val P & Swain A 2010 Gene dosage effects and transcriptional regulation Drake TA & Lusis AJ 2006 Tissue-specific expression and regulation of early mammalian adrenal cortex development. Molecular and of sexually dimorphic genes in mice. Genome Research 16 995–1004. Cellular Endocrinology 323 105–114. (https://doi.org/10.1016/j. (https://doi.org/10.1101/gr.5217506) mce.2009.12.010) Yates R, Katugampola H, Cavlan D, Cogger K, Meimaridou E, Hughes C, Val P, Martinez-Barbera JP & Swain A 2007 Adrenal development is Metherell L, Guasti L & King P 2013 Adrenocortical development, initiated by Cited2 and Wt1 through modulation of Sf-1 dosage. maintenance, and disease. Current Topics in Developmental Biology Development 134 2349–2358. (https://doi.org/10.1242/dev.004390) 106 239–312. Viger RS, Mertineit C, Trasler JM & Nemer M 1998 Transcription factor Yoshida M, Suda Y, Matsuo I, Miyamoto N, Takeda N, Kuratani S & GATA-4 is expressed in a sexually dimorphic pattern during mouse Aizawa S 1997 Emx1 and Emx2 functions in development of dorsal gonadal development and is a potent activator of the Mullerian telencephalon. Development 124 101–111. inhibiting substance promoter. Development 125 2665–2675. Zorrilla M & Yatsenko AN 2013 The genetics of infertility: current status Wainwright EN, Jorgensen JS, Kim Y, Truong V, Bagheri-Fam S, of the field. Current Genetic Medicine Reports 1 247–260. (https://doi. Davidson T, Svingen T, Fernandez-Valverde SL, McClelland KS, Taft RJ, org/10.1007/s40142-013-0027-1)

Received in final form 18 July 2018 Accepted 24 July 2018 Accepted Preprint published online 24 July 2018

https://jme.bioscientifica.com © 2019 Society for Endocrinology https://doi.org/10.1530/JME-17-0314 Published by Bioscientifica Ltd. Printed in Great Britain Downloaded from Bioscientifica.com at 09/29/2021 03:48:41AM via free access