pmc.ncbi.nlm.nih.gov

Male sex determination: insights into molecular mechanisms

Abstract

Disorders of sex development often arise from anomalies in the molecular or cellular networks that guide the differentiation of the embryonic gonad into either a testis or an ovary, two functionally distinct organs. The activation of the Y-linked gene Sry (sex-determining region Y) and its downstream target Sox9 (Sry box-containing gene 9) triggers testis differentiation by stimulating the differentiation of Sertoli cells, which then direct testis morphogenesis. Once engaged, a genetic pathway promotes the testis development while actively suppressing genes involved in ovarian development. This review focuses on the events of testis determination and the struggle to maintain male fate in the face of antagonistic pressure from the underlying female programme.

Keywords: gonadal, knockout, mice, sex determination, sex-determining region Y protein, sex disorders, sex reversal, sexual development, testis

Introduction

Perceptions of sex and sexuality pervade modern culture. However, it is important to recognize that not all members of our society fit comfortably the socially constructed ideas of masculinity and femininity. These people are likely to struggle with a variety of medical and psychosocial issues surrounding their sexuality.1, 2 It is estimated that 1.7% of all live births have a disorder of sex development (DSD).3, 4 These conditions are congenital and are characterized by chromosomal or gonadal sex that does not match outward appearance of maleness or femaleness, or anatomical sex that is in some way ambiguous or intermediate between male and female.4 Some of these conditions are associated with infertility, predisposition to gonadal tumours and/or other syndromic features.5, 6 Clearly, discovery of the underlying molecular causes of DSDs is an important goal in biomedical research.

Genomic and structure/function studies of human DSDs have revealed a number of genes as being important for sex development, while studies in the mouse have further extended our understanding of the mechanism of action of these genes; these approaches are complementary. Identifying the molecular mechanisms behind sex determination and differentiation will lead to more accurate diagnosis and prognosis, and assist in providing more informed options for psychological, endocrinological, surgical and other clinical management of DSDs, many of which remain uncharacterized at a molecular level. In a broader context, understanding the events of early testis development may also illuminate some of the underlying causes of male infertility.

In this review, we examine the molecular mechanisms behind male sex determination and differentiation, and how impairment of these mechanisms underlies a subset of human DSDs. In particular, we highlight the interplay between the molecular pathways that promote male and female development, and the role of gene dosage and phenotype sensitivity in mice and humans.

Sry and the beginnings of maleness

We each inherit an X or a Y sex chromosome from our father and an X chromosome from our mother during fertilisation. The resulting chromosomal sex (XX or XY) leads to the transformation of the embryo into a male or a female. Before gonadal sex determination in both XX and XY embryos, a bipotential gonadal primordium exists that has the potential to differentiate into either testes or ovaries. Activation of the Y-linked gene Sry (sex-determining region Y) initiates testicular development. When Sry is expressed ectopically in XX mice, the testis pathway is initiated.7 When Sry is not present, as in XX individuals, or non-functional in XY individuals, the bipotential gonads generally do not follow the testicular pathway and instead develop into ovaries.8, 9

SRY plays a role in a number of DSDs: mutation or loss of function of SRY results in complete male to female sex reversal,10, 11 whereas ectopic expression of SRY in XX individuals due to chromosomal translocation of SRY may result in female to male sex reversal. Indeed, SRY translocation is responsible for 10% of all 46,XX female to male sex reversal.12 Formation of ovotestes, where ovarian and testicular tissues coexist in the same organ, can also occur in cases of ectopic SRY activity.13, 14

SRY is a transcription factor with a DNA-binding high-mobility group box domain.15, 16 In mice, expression of Sry is both brief and carefully regulated; however, the factors controlling this burst of expression remain unknown. One of the factors postulated to play a role in activation of Sry is Wilms' tumour 1 (WT1), which can act as a transcriptional activator17, 18 or repressor.19 WT1 has two active isoforms in the gonad, with either an insertion or an omission of three amino acids, lysine (K), tyrosine (T) and serine (S), between two zinc finger motifs.20, 21 Each isoform has distinct functions during mouse testis determination. Knockout mice show that WT1+KTS, although unlikely to be directly regulating Sry expression, is required for maintenance of the gonad.20 However, this WT1 isoform has been shown to be able to transactivate the human SRY promoter in vitro.22 Conversely, WT1+KTS-null mice exhibit complete XY sex reversal, presumably due to abnormally low Sry expression.20 In agreement with the mouse model, WT1 haploinsufficiency, resulting in reduced levels of WT1+KTS, results in XY sex reversal in human patients.23 It is proposed that WT1+KTS is involved in cell-autonomous regulation of Sry in vivo, as indicated by reduced SRY levels in cells of WT1+KTS-null mouse gonads.24 It is important when interpreting these results to take into account the fact that knocking out one WT1 isoform leads to an increase in expression of the other, which may have an impact on the observed phenotype and interaction with SRY.

In addition to sufficiently early onset of expression of Sry, a threshold level of expression must be achieved for complete testis differentiation to occur. In mice, expression of Sry is initiated at 10.5 days post-coitum (d.p.c.), peaks at 11.5 d.p.c. and is extinguished by 12.5 d.p.c.25, 26, 27 Sry expression occurs in a wave-like pattern, beginning in the central region of the gonad and expanding out towards the poles.28, 29, 30, 31 In humans, SRY has a broader spatiotemporal expression profile, occurring in multiple tissues such as the adrenal and heart, and being maintained for longer in the testis, apparently through to adulthood.32 Sry/SRY is also expressed in the brain of mice and humans.32, 33, 34 In mice, SRY is expressed in a subset of nigrostriatal doperminergic neurons in the brain and appears to affect the specific motor behaviours they control.35 However, although SRY is suspected of being involved in sexual dimorphism of the brain, a specific role for SRY outside sex determination has yet to be conclusively demonstrated.

Before turning to the cellular role of SRY, it is important to discuss briefly the course of events that occur as the testis differentiates. The gonads arise from a pair of bipotential primordia known as the genital ridges. In males, differentiation of the bipotential supporting cell lineage into Sertoli cells results in organisation of the developing testis into two main compartments: the testis cords, which comprise aggregates of germ cells surrounded by a layer of Sertoli cells in turn encased by peritubular myoid cells, and the testis interstitium which includes the steroidogenic Leydig cells and the testis vasculature. The development of secondary sexual characteristics in the embryo, such as external genitalia, is directed by the testes. Thus, the morphogenesis of the bipotential gonads into testes dictates the phenotypic sex of the male individual (Figure 1).

Figure 1.

Figure 1

Overview of mouse gonadogenesis. The expression of Sry and Sox9 at 10.5–11.5 d.p.c. in the bipotential gonad initiates testis differentiation. By 13.5 d.p.c., basic testis morphology is established; the formation of testis cords, the coelomic blood vessel, and differentiation and activation of steroidogenesis in Leydig cells has occurred, and androgens are then produced by the testes. In the ovary, further differentiation is delayed. Around 13.5 d.p.c., germ cells have entered meiosis and vascularisation, and remodelling of the ovary to form germ cell cysts occurs. Later, the cortical and medullar domains begin to be established and folliculogenesis takes place. Secondary sexual characteristics include the establishment of the male and female genital tract and duct system, sex-specific brain dimorphisms and behaviours, and external genitalia. The establishment of secondary sexual characteristics involves organ-specific, regulatory gene networks. d.p.c., days post-coitum; FLC, foetal Leydig cell; Sox9, Sry box-containing gene 9; Sry/SRY, sex-determining region Y.

In mice, the first known cellular difference between the XX and XY gonad after expression of Sry is the male-specific proliferation of the epithelium at the coelomic surface of the genital ridges.36, 37 This sex-specific proliferation is thought to amplify the population of cells capable of differentiating into Sertoli cells, the first testicular cell type to differentiate,38 and is required for the formation of testis cords.36, 37, 39 Thus far, the molecular mechanism that induces coelomic epithelial proliferation is unknown. We do know a direct molecular target of SRY: the gene encoding the transcription factor Sox9 (Sry box-containing gene 9).40 Because testes develop normally in transgenic XX mice overexpressing Sox9, it appears that male-specific proliferation of the coelomic epithelium and all other aspects of foetal testis development are under the control, directly or indirectly, of SOX9.41, 42

Sox9 and the differentiation of the Sertoli cell

Sox9 is upregulated when a protein complex of SRY and steroidogenic factor 1 ((SF1) nuclear receptor subfamily 5, group A, member 1) binds to a Sox9 enhancer element known as testis-specific enhancer of Sox9 core element (TESCO).40 Like SRY, SOX9 is necessary for testis differentiation: mice lacking Sox9 undergo complete XY sex reversal,43, 44 while 75% of human patients with a heterozygous mutation in SOX9 manifest with complete or partial XY sex reversal.45, 46

The proposed mechanism for XY gonadal transcription of Sox9 during sex determination consists of three distinct phases, according to Sekido and Lovell-Badge.40 Firstly, SF1 initiates low-level transcription of Sox9 in XX and XY genital ridges. Secondly, SF1, in a complex with SRY, activates male-specific transcription of Sox9 in the male genital ridge via TESCO. High levels of SOX9 are then maintained in the XY gonad via an autoregulatory feedback loop.40, 47 Indeed, in vitro studies using SF1, SRY and SOX9 mutant proteins, modelled on clinical human mutations from 46,XY DSD patients, support this model. These proteins failed to activate the human homologue of TESCO, providing a potential mechanism by which mutations resulting in partially functional proteins can present as DSDs.48

It is believed that bipotential supporting cells cell-autonomously differentiate into Sertoli cells under the influence of SRY and SOX9: this conclusion was drawn from XX–XY chimaera studies in which it was found that, when testes formed, almost all Sertoli cells were XY, while other cell types did not exhibit a chromosomal bias.49, 50 However, some Sertoli cells were always found to be XX,49 indicating the existence of paracrine pathways by which SRY- and SOX9-positive cells can recruit additional cells (such as XX cells in the chimaera experiments, or cells that express unusually low levels of Sry in normal XY gonads) to the Sertoli fate.

Two independent mechanisms of Sertoli cell recruitment are known: fibroblast growth factor 9 (FGF9) and prostaglandin D2 (PGD2) recruitment. Kim et al.51 demonstrated using Fgf9-null mice, which exhibit XY sex reversal,52 that FGF9 is required only for the maintenance of SOX9 expression, not its initiation. However, ectopic application of FGF9 to XX gonads induces SOX9 expression.51, 53 Hiramatsu et al.54 showed that Fgf9 expression occurs in a wave emanating from the central zone of the gonad similar to Sry and Sox9. Inhibition of FGF signalling repressed the expansion of the Sox9-positive domain in the XY gonad. Furthermore, removal of the central segment or isolation of the central domain of the testis before the expansion of Fgf9 signalling resulted in failure of tubulogenesis in the anterior and posterior segments. Based on these findings, Hiramatsu et al.54 proposed a system where FGF9 was produced in the central domain of the gonad by newly specified Sertoli cells, from which it was secreted and rapidly diffused towards the gonadal poles where it recruited cells to the Sertoli fate by reinforcing the expression of Sox9. This mechanism is supported by evidence from Fgfr2-null (FGF-receptor 2) mice whose phenotype is similar to Fgf9-null mice, displaying male-to-female sex reversal and suggesting FGFR2 is the receptor for FGF9 in the XY gonad. Indeed, conditional deletion of Fgfr2 in pre-Sertoli cells shows that FGFR2 is required in pre-Sertoli cell differentiation.55

Independently, PGD2, an early product of the testis, is also able to induce Sertoli cell differentiation in vivo by amplifying SOX9 activity and canalizing the male pathway. Treatment of XX gonads with PGD2 resulted in upregulation in Sox9 and its direct downstream target Amh (the gene encoding anti-Müllerian hormone), masculinizing the XX gonad.56, 57, 58 However, Pgds (prostaglandin D2 synthase)-null testes, after a delay in Sertoli cell differentiation, develop normally, indicating that this mechanism is a nonessential backup system for Sertoli cell recruitment.56 The requirement of theses backup systems and the need to continually reinforce the male programme may stem from the weak but critical role of SRY and the need to actively suppress the underlying female programme. Existence of these recruitment mechanisms ensures that as many cells as required are pulled into the Sertoli fate to allow successful differentiation of the gonad.

The role of the testis vasculature

Vascular patterning in the gonad is a sex-specific process.59 Testis vasculature is formed by migration of endothelial cells into the developing testes.60, 61 By 12.5 d.p.c., a prominent artery known as the coelomic blood vessel can be seen along the anterior–posterior length of the testis, in addition to extensive microvasculature. Ectopic coelomic vessel-like structures were observed in gonads of XX mice mutant for Rspo1 (R-spondin homologue 1), Wnt4 (wingless-related MMTV integration site 4), Fst (follistatin) and Ctnnb1 (catenin (cadherin-associated protein), beta 1).62, 63, 64, 65, 66 Rspo1 is a regulator of WNT4 signalling which involves Ctnnb1, while Fst is downstream of WNT4,67 implicating the WNT signalling pathway in vessel formation and patterning. Additionally, overexpression of Wnt4 disrupts normal testis vasculature, indicating that WNT4 inhibits formation of gonad vasculature.68 Notably, where testis vasculature is disrupted, as in the WNT4 overexpressing mice, Sertoli and foetal Leydig cells still differentiate.65, 68

Recently, it has been discovered that the vascularisation of the testis plays an important instructive role in testis cord formation.60, 61 When endothelial migration was suppressed in testes by blocking vascular endothelial growth factors with VEGF-Trap or by using an antibody against vascular endothelial cadherin, testis cord morphogenesis was impaired.60, 69 Antagonizing vessel maturation also reduced proliferation of interstitial mesenchymal cells that appear to segregate the precursor territories for testis cords; this proliferation could be rescued by the addition of platelet derived growth factor isoform BB (PDGF-BB).61, 69, 70 However, the mechanisms governing testis vascularisation and cord segregation are still unclear.

Leydig cells: the key to phenotypic masculinisation

Foetal Leydig cells (FLCs) produce steroid hormones that reinforce male-specific differentiation of the testis (for review, see Ref. 71). The FLC populations in humans and mice are similar during foetal life,72, 73 although observation of the induction of steroidogenesis largely limited to steroid level quantification. The origins of FLCs in humans remain unclear. In the mouse, FLCs arise by about 12.5 d.p.c. (for review, see Ref. 74), and recent work involving cell lineage tracing and live imaging suggests that they arise from multiple origins including the coelomic epithelium and the gonad/mesonephros border.53 Several pathways and molecules have been implicated in their differentiation and maintenance. SF1 marks presteroidogenic and pre-Sertoli cells in the developing genital ridge75, 76 and acts as a key regulator of genes encoding steroid hydroxylases, which later distinguish FLCs.77 Members of the hedgehog signalling pathway, desert hedgehog (DHH), which is secreted by Sertoli cells, and its receptor patched homologue 1 (PTCH1), which is expressed by the interstitium, have been shown to be positive regulators of FLC differentiation.78 Dhh-null mice have a FLC differentiation defect.78 Human patients with mutations in DHH present with mixed, partial or pure gonadal dysgenesis through to seemingly unaffected carriers.79, 80, 81 In addition, chemical inhibition of hedgehog signalling at 11.5 d.p.c. completely abolished expression of steroidogenic enzymes, confirming that DHH/PTCH1 signalling is essential for FLC differentiation.78 Gli1 (glioma-associated oncogene family zinc finger 1) and Gli2 (glioma-associated oncogene family zinc finger 2) are downstream targets of hedgehog signalling and are expressed exclusively in the testis interstitium in a manner similar to PTCH1.82 However, Gli1- and Gli2-null mice display normal FLC differentiation, perhaps indicating functional redundancy between the GLI factors in FLCs.82 Ectopic activation of the hedgehog signalling pathway in SF1-positive ovarian cells is sufficient to differentiate these cells into functional FLCs within an ovarian environment.83 These ectopic cells upregulated SF1 and were able to partially masculinize the phenotype of the XX embryo.83 Additionally, Pdgfr-α (platelet-derived growth factor receptor, alpha polypeptide) and Arx (Aristaless-related homeobox) have been identified as being critical for FLC differentiation in knockout mouse models.84, 85

Germ cells: the origin of sperm

Germ cells are the precursors of oocytes and spermatozoa in the foetal gonad. The sexual fate of the germ cell is determined by signalling factors that the germ cells are exposed to upon entry to the gonad, rather than by their chromosomal constitution.58, 86, 87, 88, 89 Much of what is known about the origin and regulation of the germ cells is derived from studies in mice, as discussed below.

In an ovary, germ cells must enter meiosis during foetal life if they are to initiate oogenesis correctly; conversely, meiosis must be avoided in male germ cells in the foetus if they are to embark on the spermatogenic pathway. The interplay between FGF9 and retinoic acid (RA) appears to be key to the correct specification of the germ cells in the mouse: meiosis is induced by RA in the foetal ovary and inhibited by FGF9, which is secreted by Sertoli cells, in the foetal testes.86, 87, 88, 90 In the developing testis, meiosis is avoided because RA is degraded by the P450 enzyme CYP26B1 (cytochrome P450, family 26, subfamily b, polypeptide 1).91 Thus, CYP26B1 acts to suppress meiosis indirectly by the removal of RA, while FGF9 directly suppresses meiosis and acts to maintain pluripotency.86, 87, 88 This mechanism is supported by in vivo evidence from Cyp26B1-null mice where degradation of RA does not occur in XY gonads, resulting in upregulation of RA-responsive Stra8 (stimulated by retinoic acid gene 8) and germ cell entry into meiosis.87, 91

Recently, a double-knockout of Aldh1a2/Aldha1a3 (aldehyde dehydrogenase family 1, subfamily A2/A3), genes encoding key synthesizers of RA in the mesonephros, demonstrated that some meiosis still occurred in the foetal ovary.92 These data indicate that either RA does not drive meiosis or, more likely, that there is an additional source of RA that remains in these mice. More in-depth analysis of this model will be required to clarify this point. Regardless, a strong antagonism exists between meiosis-promoting (female) factors and meiosis-suppressing (male) factors that push the resident germ cells into their respective fates.

Relatively, little is known about whether these mechanisms are used in humans. Culture experiments demonstrate that the RA initiates meiosis in the human ovary and can upregulate STRA8.93, 94 However, it appears that the human gonad has the capacity to produce RA, evidenced by the strong expression of ALDHA1A1 (aldehyde dehydrogenase family 1, subfamily A1) in the ovary around the time of meiosis initiation.93, 94 Most striking is the apparent lack of CYP26B1 expression in the foetal human testes and the expression of RA receptors, indicating that the testes may be exposed to, and may be able to respond to, RA, unlike the situation in the mouse.93, 95

Male germ cells are fated to enter G1/G0 arrest in the foetal testes.89 Retinoblastoma 1 is a cell cycle regulator necessary for male germ cells to enter arrest at the appropriate time. In XY retinoblastoma 1-null mice, the germ cell population fails to enter G1/G0 arrest appropriately.96 To compensate cell cycle suppressors, cyclin-dependent kinase inhibitors 1b and 2b are upregulated and after a delay can induce arrest.96

Ovotestes: what they reveal about male–female antagonism in the embryo

The study of ovotestes in mouse models has provided numerous insights into the antagonism between the male and female pathways during sex determination. When the Y chromosome derived from Mus poschiavinus, YPOS, is backcrossed to a C57/BL6 (B6) background, varying degrees of sex reversal are observed in the XY progeny.97 This phenomenon is thought to be due to defective interaction between Sry on YPOS and autosomal sex-determining genes in B6.98 Detailed expression studies have shown that a delay in the commencement of Sry expression, and subsequently, Sox9 expression is the likely cause of B6-YPOS partial sex reversal.98, 99, 100 Wilhelm et al.30 found that although Sry was expressed throughout the genital ridge in B6-YPOS mice, the upregulation of Sox9 and activation of downstream testis differentiation pathways only took place in the central zone where Sry expression was initiated.25, 28, 29, 31 These findings indicate that expression of Sry in the poles, which in ovotestes differentiate into ovarian tissue, does not reach the required expression threshold early enough. This failure to upregulate Sox9 in pre-Sertoli cells allows the expression of key ovarian differentiation genes and the engagement of the ovarian programme. Therefore, male–female antagonism underlies the successful differentiation of the gonad. When the balance of factors is altered, even slightly, the underlying battle between the testicular and ovarian fate is revealed.

Molecular balancing acts: examples of antagonism between male and female pathways

In order to understand the molecular mechanisms behind male sex determination, we must also understand what is occurring molecularly in the antagonistic female programme at the time of sex determination. It has become clear that the ovarian programme, although it is considered the ‘default', is an active genetic programme in its own right.101 The antagonism between the male and female pathways has been illustrated using genetic approaches (Figure 2). In an early study of male–female antagonism, the male pathway was suppressed by knocking out Fgf9; as a result, the female pathway was promoted, indicated by the upregulation of WNT4.51 Conversely, when the female pathway was suppressed by knocking out Wnt4, the male pathway was stimulated.102 This phenomenon correlates to cases in DSD patients where WNT4 loss or mutation results in XX masculinisation and duplication results in XY feminisation.68, 103 This male–female antagonism is further supported by ex vivo work. Treatment of XX gonads with ectopic FGF9 suppressed normal WNT4 expression and induced ectopic upregulation of Sox9.51 Thus, readouts of the male pathway, SOX9 and FGF9 can increase in response to a weakening of the female programme and vice versa.

Figure 2.

Figure 2

Model of molecular interactions and proposed levels of antagonism in testis and ovary. For details see text. In the embryonic XY gonad, Sry is activated via a mechanism involving WT1 (indirect regulation illustrated by dotted arrow/line). SRY then directly upregulates Sox9, direct regulation illustrated by solid arrow/line) which maintains its own expression; positive feedback loops exists between PGD2/SOX9 and FGF9/SOX9. In the XX gonad, FOXL2 is active as are RSPO1, β-catenin and WNT4, which is required for expression of Fst. During embryonic development, antagonism is though to exist between FGF9/WNT4, SOX9/WNT4 and SOX9/FOXL2. Postnatally, antagonism exists between SOX9/FOXL2 and DMRT1/FOXL2. DAX1, Nr0b1, nuclear receptor subfamily 0, group B, member 1; DMRT1, doublesex and mab-3-related transcription factor 1; FGF9, fibroblast growth factor 9; FOG2, ‘Friend of Gata1' type 2; FOXL2, forkhead box L2; Fst/FST, follistatin; GATA4, GATA-binding protein 4; PGD2, prostaglandin D2; RSPO1, R-spondin homologue 1; SF1, steroidogenic factor 1; Sox9/SOX9, Sry box-containing gene 9; Sry/SRY, sex-determining region Y; WNT4, wingless-related MMTV integration site 4; WT1, Wilms' tumour 1.

Recent studies have shown that the balance between the antagonistic testicular and ovarian differentiation programmes is also important postnatally. Gonad-specific transcription factors doublesex and mab-3-related transcription factor 1 (DMRT1) and forkhead box L2 (FOXL2) have roles in maintenance of the testes and ovaries respectively.104 In humans, DMRT1 hemizygosity may result in hypogonadism, often with streak gonads.105 FOXL2/FoxL2 is a key ovarian marker, and its mutation is associated with premature ovarian failure in human patients and knockout mice.104, 106, 107 Dmrt1-null males were found to have numerous FOXL2-positive cells in the seminiferous tubules 4 months after birth.108 A Sertoli cell-specific knockout of Dmrt1 confirmed that loss of DMRT1 in Sertoli cells and not germ cells allows ectopic FOXL2 expression.108 However, FOXL2 may also be able to repress DMRT1, as shown by strong upregulation of DMRT1, when FOXL2 is ablated postnatally,109 suggesting the existence of a mutual antagonism necessary for maintaining sex differentiation throughout life.

FOXL2 has also been shown to antagonize SOX9 in the adult ovary and is postulated to also play a role in the embryonic testes. Conditional deletion of FoxL2 at 8 weeks resulted in transdifferentiation of granulosa cells in the adult ovary to Sertoli-like cells expressing SOX9 even in the presence of oocytes and theca cells, the steroidogenic cells of the ovary, differentiated into Leydig-like cells.109 In the adult ovary, it was demonstrated using chromatin immunoprecipitation that FOXL2 can repress TESCO activity,109 providing a possible mechanism for the role of FOXL2 in maintaining ovarian function postnatally. FoxL2 is also upregulated in XY embryonic gonads of Sox9-conditional null mice indicating that a similar mechanism exists during embryonic sex differentiation.43 However, importantly, expression of male and female factors in the gonad is exclusive, as observed in the ovotestes,30 such that FOXL2 and SOX9 are never coexpressed in the same cell. Nevertheless, while the factors mentioned in the above section are responsive to each other's loss, direct interaction and antagonism between any of these factors is yet to be demonstrated.

SF1: highlighting the role of gene dosage differences in the mouse and human

SF1 is a striking example of a factor whose effects on gonadal phenotype are sensitive to dosage and genetic background.75 Sf1-null mice undergo early gonadal and adrenal development, but the organs regress by 11.5 d.p.c.75, 110 Transgenic expression of SF1 in Sf1-null mice was able to rescue gonad development, but not adrenal development.111 Additionally, Sf1-haploinsufficient mice have disrupted adrenal development but develop testes normally.112 Together, these results indicate that SF1 is involved in gonad and adrenal organ maintenance and that, in the mouse, the adrenal is more sensitive to Sf1 dosage than the gonad. However, there is evidence that the reverse is true in humans. Two patients with heterozygous mutations in SF1 have been shown to exhibit gonadal dysgenesis but normal adrenal function, indicating that, in humans, testis development is more sensitive to Sf1 dosage than adrenal gland development.113, 114

DAX1: a question of gene dosage and genetic background

Dax1 (Nr0b1, nuclear receptor subfamily 0, group B, member 1) expression in the mouse indicates a role in early sex determination, with Dax1 being expressed in both sexes initially before being downregulated in the testes.31, 115 Duplications of chromosomal region Xp21.2–21.1, which includes DAX1, are sufficient to impair testis differentiation in 46,XY human patients and induce male-to-female sex reversal (dosage-sensitive sex reversal).116, 117, 118 These data imply that DAX1 acts as an antitestis gene, antagonizing the action of SRY. Similarly, overexpression of Dax1 in mice can induce male-to-female sex reversal. However, only mice highly overexpressing Dax1 and which also have a weakened male sex-determining pathway driven by a YPOS chromosome, show complete male-to-female sex reversal.31 A common interpretation of these data is that mice and humans may have different sensitivity thresholds for DAX1 dosage.

An alternative explanation is that genetic background and the presence of genetic modifiers can explain whether or not gonad sex reversal occurs. Genetic background is a key determinant in the penetrance of a number of sex reversal phenotypes, with heterozygous mutants often displaying different phenotypes depending on their genetic background. Bouma et al.119 compared sex development in Gata4 (GATA-binding protein 4)120, 121 and Fog2 (‘Friend of Gata1' type 2; Zfpm2, zinc finger protein, multitype 2)120, 121 heterozygous mutants on B6/YAKR, B6, D2/YAKR and D2 backgrounds, which have decreasing sensitivity to sex reversal. They found that ovaries or ovotestes developed in these mutants, but only on the B6/YAKR background. All other strains developed testes normally. These studies demonstrate the importance of genetic background, and also clearly indicate a role for GATA4 and FOG2 in foetal testis development. A similar background dependance was observed in the generation of sex-reversed Dax1 mutant mice.31, 122, 123, 124 Given that genetic background and the presence of genetic modifiers can explain whether or not gonad sex reversal occurs, it follows that effects attributed to differences in dosage sensitivity between humans and mice might also be explained by genetic background.

Given the expectation that Dax1 acts as an antitestis gene, deletion of Dax1 in mice produced a surprising result: ovaries formed normally in Dax1-null XX mice, but in XY mice with a deletion of Dax1, testis cord formation was abnormal.123 Moreover, in XY Dax1−/−/YPOS mice, complete male-to-female sex reversal was observed.124 Subsequent studies revealed the cause of the male-to-female sex reversal: Sox9 is not upregulated in XY Dax1-null mice, despite normal expression of Sry.125 Nonetheless, strong overexpression of Sry in XYPOS Dax1-null mice was able to upregulate Sox9, correcting testis development and producing fertile males.125 These results are somewhat in accord with findings in humans, with respect to DAX1 mutations. 46,XY patients with deletion of, or mutations in, DAX1, exhibit the male phenotype, but have hypogonadotrophic hypogonadism and disorganized testis cords, indicating that DAX1 is not essential for the initial stages of human gonad development, but is required for testis development.117, 124, 125 As there is evidence that DAX1 can work in both ‘antitestis' and ‘protestis' capacities, it is possible that correct function in the testis occurs only within a ‘window' of activity involving two concentration thresholds. In this scenario, DAX1 activity beyond an upper threshold, for example, in cases of DAX1 duplication, may act to antagonize testis differentiation, while DAX1 activity below a lower threshold, for example, in cases of mutation, may allow ovarian differentiation to occur.126

Conclusions and perspectives

From the above discussion, it is clear that initial specification of Sertoli cells is a result of SRY expression, with the effects of SRY mediated largely if not entirely by SOX9. A complete understanding of male sex determination and testis morphogenesis, therefore, largely depends on a deeper understanding of the molecular and cellular roles of SOX9, and therefore, on the discovery and characterisation of all transcriptional targets of SOX9. Further, it remains to be clarified how the Sertoli cell directs events during morphogenesis; that is, what signalling molecules are produced by these cells and how they influence the differentiation of the other testicular cell lineages. How the other cell lineages, once masculinized, then contribute to testis morphogenesis is also a question that requires further investigation. A greater understanding of the molecular interactions involved in the process of testis differentiation will provide new avenues for DSD diagnosis and management.

Commitment to the male fate and then maintenance of that fate is achieved only by overcoming the progress of the female pathway, and antagonistic interplay is seen during foetal life, as well as postnatally. Mutual antagonism is likely to be facilitated by transcription factors such as SOX9 and FOXL2, but assays to determine the molecular interactions that underpin these antagonistic relationships have yet to be completed. Understanding what factors facilitate antagonism between the male and female programmes will lead to a greater understanding of what pushes individuals into or out of the two typical sex phenotypes specified by XX and XY chromosomes.

Additionally, as demonstrated here, sex-determining genes often respond in a dosage-dependent manner that may also be influenced by genetic background. As a result, human DSDs and mouse models may not always phenocopy each other, highlighting the need for a collaborative approach to DSD identification and diagnosis between researchers and clinicians. Human patient cases demonstrate, especially, that gene dosage and genetic background can be important factors in phenotype severity. However, despite this, the investigation of gene dosage effects and the effects of genetic background on sex determining genes in the mouse is a question that few researchers have addressed and that warrants further detailed investigation. Tools, such as inducible knockout and single-copy transgenesis strategies, are now available to study these phenomena in the mouse. Thus, better integration of lessons from both human cases and mouse models must be a priority. Finally, it is clear that integration of clinical findings and mouse models will contribute to a better understanding not only of the causes of human DSDs but also of the basic biology of sex determination in the male.

Abbreviations: DSD, disorder of sex development; DNA, deoxyribonucleic acid; HMG, high-mobility group-box; WT1/WT1/Wt1, Wilms Tumor 1; K, lysine; T, tyrosine; S, serine; Sry, sex determining region of Chr Y; SOX9/SOX9/Sox9, SRY-box containing gene 9; SF1/SF1/Sf1 (Nr5a1), steroidogenic factor 1 (nuclear receptor subfamily 5, group A, member 1); TESCO, testis-specific enhancer of Sox9 core element; FGF9/FGF9/Fgf9, fibroblast growth factor 9; PDG2/PDG2/Pdg2, prostaglandin D2 synthase; FGF, fibroblast growth factor; FGFR2/FGFR2/Fgfr2, Fgf-receptor 2; AMH/AMH/Amh, anti-Mullerian hormone; PGDS/PGDS/Pgds, prostaglandin D2 synthase; dpc, days post coitum; WNT4/WNT4/Wnt4, wingless-related MMTV integration site 4; FLC, fetal Leydig cell; DHH/DHH/Dhh, desert hedgehog; PTCH1/PTCH1/Ptch1, patched homolog 1; GLI1/2/GLI1/2/Gli1/2, glioma-associated oncogene family zinc finger 1/2; PDGFR-α/PDGFR-α/PDGFR-α, platelet derived growth factor receptor, alpha polypeptide; ARX/ARX/Arx, aristaless related homeobox; RA, retinoic acid; STRA8/STRA8/Stra8, stimulated by retinoic acid gene 8; RB1/RB1/Rb1, retinoblastoma 1; Cdkn1b/2b/Cdkn1b/2b/Cdkn1b/2b, cyclin dependent kinase inhibitors 1b/2b; CYP26B1/CYP26B1/Cyp26b1, cytochrome P450, family 26, subfamily b, polypeptide 1; Aldh1a1/2/3/Aldh1a1/2/3/Aldh1a1/2/3, aldehyde dehydrogenase family 1, subfamily A1/A2/A3; YPOS, Y chromosome derived from Mus poschiavinus; β-catenin/CTTNB1/Ctnnb1, catenin (cadherin associated protein), beta 1; FOXL2/FOXL2/FoxL2, forkhead box L2; DMRT1/DMRT1/Dmrt1, doublesex and mab-3 related transcription factor 1; FOG2/FOG2/Fog2, Zfpm2, zinc finger protein, multitype 2; GATA4/GATA4/Gata4, GATA binding protein 4; DAX1/DAX1/Dax1, Nr0b1, nuclear receptor subfamily 0, group B, member 1; FST/FST/Fst, follistatin; RSPO1/RSPO1/Rspo1, R-spondin homolog; PDGF-BB, platelet derived growth factor isoform BB.

Acknowledgments

We thank Elanor Wainwright for critical reading of this manuscript. This work was supported by research grants from the Australian Research Council (ARC) and National Health and Medical Research Council of Australia. PK is a Federation Fellow of the ARC.

The authors declare no competing financial interests.

References

  1. Pasterski V, Prentice P, Hughes IA. Impact of the consensus statement and the new DSD classification system. Best Pract Res Clin Endocrinol Metab. 2010;24:187–95. doi: 10.1016/j.beem.2009.11.004. [DOI] [PubMed] [Google Scholar]
  2. Gupta D, Bhardwaj M, Sharma S, Ammini A, Gupta D. Long-term psychosocial adjustments, satisfaction related to gender and the family equations in disorders of sexual differentiation with male sex assignment. Pediatr Surg Int. 2010;26:955–8. doi: 10.1007/s00383-010-2661-y. [DOI] [PubMed] [Google Scholar]
  3. Blackless M, Charuvastra A, Derryck A, Fausto-Sterling A, Lauzanne K, et al. How sexually dimorphic are we? Review and synthesis. Am J Hum Biol. 2000;12:151–66. doi: 10.1002/(SICI)1520-6300(200003/04)12:2<151::AID-AJHB1>3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  4. Hughes IA, Houk C, Ahmed SF, Lee PA. LWPES1 Consensus Group. et al Consensus statement on management of intersex disorders. Arch Dis Child. 2006;91:554–63. doi: 10.1136/adc.2006.098319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Looijenga LH, Hersmus R, de Leeuw BH, Stoop H, Cools M, et al. Gonadal tumours and DSD. Best Pract Res Clin Endocrinol Metab. 2010;24:291–310. doi: 10.1016/j.beem.2009.10.002. [DOI] [PubMed] [Google Scholar]
  6. Woodhouse CR. Prospects for fertility in patients born with genitourinary anomalies. J Urol. 2001;165:2354–60. doi: 10.1016/S0022-5347(05)66202-7. [DOI] [PubMed] [Google Scholar]
  7. Koopman P, Gubbay J, Vivian N, Goodfellow P, Lovell-Badge R. Male development of chromosomally female mice transgenic for Sry. Nature. 1991;351:117–21. doi: 10.1038/351117a0. [DOI] [PubMed] [Google Scholar]
  8. Gubbay J, Collignon J, Koopman P, Capel B, Economou A, et al. A gene mapping to the sex-determining region of the mouse Y chromosome is a member of a novel family of embryonically expressed genes. Nature. 1990;346:245–50. doi: 10.1038/346245a0. [DOI] [PubMed] [Google Scholar]
  9. Lovell-Badge R, Robertson E. XY female mice resulting from a heritable mutation in the primary testis-determining gene, Tdy. Development. 1990;109:635–46. doi: 10.1242/dev.109.3.635. [DOI] [PubMed] [Google Scholar]
  10. Jager RJ, Anvret M, Hall K, Scherer G. A human XY female with a frame shift mutation in the candidate testis-determining gene SRY. Nature. 1990;348:452–4. doi: 10.1038/348452a0. [DOI] [PubMed] [Google Scholar]
  11. Maier EM, Leitner C, Lohrs U, Kuhnle U. True hermaphroditism in an XY individual due to a familial point mutation of the SRY gene. J Pediatr Endocrinol Metab. 2003;16:575–80. doi: 10.1515/jpem.2003.16.4.575. [DOI] [PubMed] [Google Scholar]
  12. Nieto K, Pena R, Palma I, Dorantes LM, Erana L, et al. 45,X/47,XXX/47,XX, del(Y)(p?)/46,XX mosaicism causing true hermaphroditism. Am J Med Genet A. 2004;130A:311–4. doi: 10.1002/ajmg.a.30258. [DOI] [PubMed] [Google Scholar]
  13. Margarit E, Coll MD, Oliva R, Gomez D, Soler A, et al. SRY gene transferred to the long arm of the X chromosome in a Y-positive XX true hermaphrodite. Am J Med Genet. 2000;90:25–8. doi: 10.1002/(sici)1096-8628(20000103)90:1<25::aid-ajmg5>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  14. Sharp A, Kusz K, Jaruzelska J, Tapper W, Szarras-Czapnik M, et al. Variability of sexual phenotype in 46,XX(SRY+) patients: the influence of spreading X inactivation versus position effects. J Med Genet. 2005;42:420–7. doi: 10.1136/jmg.2004.022053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Polanco JC, Koopman P. Sry and the hesitant beginnings of male development. Dev Biol. 2007;302:13–24. doi: 10.1016/j.ydbio.2006.08.049. [DOI] [PubMed] [Google Scholar]
  16. Kashimada K, Koopman P. Sry: the master switch in mammalian sex determination. Development. 2010;137:3921–30. doi: 10.1242/dev.048983. [DOI] [PubMed] [Google Scholar]
  17. Lee SB, Huang K, Palmer R, Truong VB, Herzlinger D, et al. The Wilms tumor suppressor WT1 encodes a transcriptional activator of amphiregulin. Cell. 1999;98:663–73. doi: 10.1016/s0092-8674(00)80053-7. [DOI] [PubMed] [Google Scholar]
  18. Shimamura R, Fraizer GC, Trapman J, Lau YfC, Saunders GF. The Wilms' tumor gene WT1 can regulate genes involved in sex determination and differentiation: SRY, Mullerian-inhibiting substance, and the androgen receptor. Clin Cancer Res. 1997;3:2571–80. [PubMed] [Google Scholar]
  19. Hewitt SM, Fraizer GC, Wu YJ, Rauscher FJ, 3rd, Saunders GF. Differential function of Wilms' tumor gene WT1 splice isoforms in transcriptional regulation. J Biol Chem. 1996;271:8588–92. doi: 10.1074/jbc.271.15.8588. [DOI] [PubMed] [Google Scholar]
  20. Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, et al. Two Splice variants of the Wilms' tumor 1 gene have distinct functions during sex determination and nephron formation. Cell. 2001;106:319–29. doi: 10.1016/s0092-8674(01)00453-6. [DOI] [PubMed] [Google Scholar]
  21. Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM, et al. Alternative splicing and genomic structure of the Wilms tumor gene WT1. Proc Natl Acad Sci USA. 1991;88:9618–22. doi: 10.1073/pnas.88.21.9618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Hossain A, Saunders GF. The human sex-determining gene SRY is a direct target of WT1. J Biol Chem. 2001;276:16817–23. doi: 10.1074/jbc.M009056200. [DOI] [PubMed] [Google Scholar]
  23. Barbaux S, Niaudet P, Gubler MC, Grunfeld JP, Jaubert F, et al. Donor splice-site mutations in WT1 are responsible for Frasier syndrome. Nat Genet. 1997;17:467–70. doi: 10.1038/ng1297-467. [DOI] [PubMed] [Google Scholar]
  24. Bradford ST, Wilhelm D, Bandiera R, Vidal V, Schedl A, et al. A cell-autonomous role for WT1 in regulating Sry in vivo. Hum Mol Genet. 2009;18:3429–38. doi: 10.1093/hmg/ddp283. [DOI] [PubMed] [Google Scholar]
  25. Hacker A, Capel B, Goodfellow P, Lovell-Badge R. Expression of Sry, the mouse sex determining gene. Development. 1995;121:1603–14. doi: 10.1242/dev.121.6.1603. [DOI] [PubMed] [Google Scholar]
  26. Jeske YW, Bowles J, Greenfield A, Koopman P. Expression of a linear Sry transcript in the mouse genital ridge. Nat Genet. 1995;10:480–2. doi: 10.1038/ng0895-480. [DOI] [PubMed] [Google Scholar]
  27. Koopman P, Munsterberg A, Capel B, Vivian N, Lovell-Badge R. Expression of a candidate sex-determining gene during mouse testis differentiation. Nature. 1990;348:450–2. doi: 10.1038/348450a0. [DOI] [PubMed] [Google Scholar]
  28. Albrecht KH, Eicher EM. Evidence that Sry is expressed in pre-Sertoli cells and Sertoli and granulosa cells have a common precursor. Dev Biol. 2001;240:92–107. doi: 10.1006/dbio.2001.0438. [DOI] [PubMed] [Google Scholar]
  29. Bullejos M, Koopman P. Spatially dynamic expression of Sry in mouse genital ridges. Dev Dyn. 2001;221:201–5. doi: 10.1002/dvdy.1134. [DOI] [PubMed] [Google Scholar]
  30. Wilhelm D, Washburn LL, Truong V, Fellous M, Eicher EM, et al. Antagonism of the testis- and ovary-determining pathways during ovotestis development in mice. Mech Dev. 2009;126:324–36. doi: 10.1016/j.mod.2009.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Swain A, Narvaez V, Burgoyne P, Camerino G, Lovell-Badge R. Dax1 antagonizes Sry action in mammalian sex determination. Nature. 1998;391:761–7. doi: 10.1038/35799. [DOI] [PubMed] [Google Scholar]
  32. Clepet C, Schafer AJ, Sinclair AH, Palmer MS, Lovell-Badge R, et al. The human SRY transcript. Hum Mol Genet. 1993;2:2007–12. doi: 10.1093/hmg/2.12.2007. [DOI] [PubMed] [Google Scholar]
  33. Lahr G, Maxson SC, Mayer A, Just W, Pilgrim C, et al. Transcription of the Y chromosomal gene, Sry, in adult mouse brain. Brain Res Mol Brain Res. 1995;33:179–82. doi: 10.1016/0169-328x(95)00136-g. [DOI] [PubMed] [Google Scholar]
  34. Mayer A, Lahr G, Swaab DF, Pilgrim C, Reisert I. The Y-chromosomal genes SRY and ZFY are transcribed in adult human brain. Neurogenetics. 1998;1:281–8. doi: 10.1007/s100480050042. [DOI] [PubMed] [Google Scholar]
  35. Dewing P, Chiang CW, Sinchak K, Sim H, Fernagut PO, et al. Direct regulation of adult brain function by the male-specific factor SRY. Curr Biol. 2006;16:415–20. doi: 10.1016/j.cub.2006.01.017. [DOI] [PubMed] [Google Scholar]
  36. Schmahl J, Eicher EM, Washburn LL, Capel B. Sry induces cell proliferation in the mouse gonad. Development. 2000;127:65–73. doi: 10.1242/dev.127.1.65. [DOI] [PubMed] [Google Scholar]
  37. Karl J, Capel B. Sertoli cells of the mouse testis originate from the coelomic epithelium. Dev Biol. 1998;203:323–33. doi: 10.1006/dbio.1998.9068. [DOI] [PubMed] [Google Scholar]
  38. Magre S, Jost A. The initial phases of testicular organogenesis in the rat. An electron microscopy study. Arch Anat Microsc Morphol Exp. 1980;69:297–318. [PubMed] [Google Scholar]
  39. Schmahl J, Capel B. Cell proliferation is necessary for the determination of male fate in the gonad. Dev Biol. 2003;258:264–76. doi: 10.1016/s0012-1606(03)00122-2. [DOI] [PubMed] [Google Scholar]
  40. Sekido R, Lovell-Badge R. Sex determination involves synergistic action of SRY and SF1 on a specific Sox9 enhancer. Nature. 2008;453:930–4. doi: 10.1038/nature06944. [DOI] [PubMed] [Google Scholar]
  41. Vidal VP, Chaboissier MC, de Rooij DG, Schedl A. Sox9 induces testis development in XX transgenic mice. Nat Genet. 2001;28:216–7. doi: 10.1038/90046. [DOI] [PubMed] [Google Scholar]
  42. Bishop CE, Whitworth DJ, Qin Y, Agoulnik AI, Agoulnik IU, et al. A transgenic insertion upstream of Sox9 is associated with dominant XX sex reversal in the mouse. Nat Genet. 2000;26:490–4. doi: 10.1038/82652. [DOI] [PubMed] [Google Scholar]
  43. Barrionuevo F, Bagheri-Fam S, Klattig J, Kist R, Taketo MM, et al. Homozygous inactivation of Sox9 causes complete XY sex reversal in mice. Biol Reprod. 2006;74:195–201. doi: 10.1095/biolreprod.105.045930. [DOI] [PubMed] [Google Scholar]
  44. Chaboissier MC, Kobayashi A, Vidal VI, Latzkendorf S, van de Kant HJ, et al. Functional analysis of Sox8 and Sox9 during sex determination in the mouse. Development. 2004;131:1891–901. doi: 10.1242/dev.01087. [DOI] [PubMed] [Google Scholar]
  45. Foster JW, Dominguez-Steglich MA, Guioli S, Kwok C, Weller PA, et al. Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 1994;372:525–30. doi: 10.1038/372525a0. [DOI] [PubMed] [Google Scholar]
  46. Wagner T, Wirth J, Meyer J, Zabel B, Held M, et al. Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell. 1994;79:1111–20. doi: 10.1016/0092-8674(94)90041-8. [DOI] [PubMed] [Google Scholar]
  47. Sekido R, Lovell-Badge R. Sex determination and SRY: down to a wink and a nudge. Trends Genet. 2009;25:19–29. doi: 10.1016/j.tig.2008.10.008. [DOI] [PubMed] [Google Scholar]
  48. Knower KC, Kelly S, Ludbrook LM, Bagheri-Fam S, Sim H, et al. Failure of SOX9 regulation in 46XY disorders of sex development with SRY, SOX9 and SF1 mutations. PLoS ONE. 2011;6:e17751. doi: 10.1371/journal.pone.0017751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Palmer SJ, Burgoyne PS. In situ analysis of fetal, prepuberal and adult XX–XY chimaeric mouse testes: sertoli cells are predominantly, but not exclusively, XY. Development. 1991;112:265–8. doi: 10.1242/dev.112.1.265. [DOI] [PubMed] [Google Scholar]
  50. Patek CE, Kerr JB, Gosden RG, Jones KW, Hardy K, et al. Sex chimaerism, fertility and sex determination in the mouse. Development. 1991;113:311–25. doi: 10.1242/dev.113.1.311. [DOI] [PubMed] [Google Scholar]
  51. Kim Y, Kobayashi A, Sekido R, DiNapoli L, Brennan J, et al. Fgf9 and Wnt4 act as antagonistic signals to regulate mammalian sex determination. PLoS Biol. 2006;4:e187. doi: 10.1371/journal.pbio.0040187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Colvin JS, Green RP, Schmahl J, Capel B, Ornitz DM. Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell. 2001;104:875–89. doi: 10.1016/s0092-8674(01)00284-7. [DOI] [PubMed] [Google Scholar]
  53. DeFalco T, Takahashi S, Capel B. Two distinct origins for Leydig cell progenitors in the fetal testis. Dev Biol. 2011;352:14–26. doi: 10.1016/j.ydbio.2011.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hiramatsu R, Harikae K, Tsunekawa N, Kurohmaru M, Matsuo I, et al. FGF signaling directs a center-to-pole expansion of tubulogenesis in mouse testis differentiation. Development. 2010;137:303–12. doi: 10.1242/dev.040519. [DOI] [PubMed] [Google Scholar]
  55. Kim Y, Bingham N, Sekido R, Parker KL, Lovell-Badge R, et al. Fibroblast growth factor receptor 2 regulates proliferation and Sertoli differentiation during male sex determination. Proc Natl Acad Sci USA. 2007;104:16558–63. doi: 10.1073/pnas.0702581104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Moniot B, Declosmenil F, Barrionuevo F, Scherer G, Aritake K, et al. The PGD2 pathway, independently of FGF9, amplifies SOX9 activity in Sertoli cells during male sexual differentiation. Development. 2009;136:1813–21. doi: 10.1242/dev.032631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Wilhelm D, Martinson F, Bradford S, Wilson MJ, Combes AN, et al. Sertoli cell differentiation is induced both cell-autonomously and through prostaglandin signaling during mammalian sex determination. Dev Biol. 2005;287:111–24. doi: 10.1016/j.ydbio.2005.08.039. [DOI] [PubMed] [Google Scholar]
  58. Adams IR, McLaren A. Sexually dimorphic development of mouse primordial germ cells: switching from oogenesis to spermatogenesis. Development. 2002;129:1155–64. doi: 10.1242/dev.129.5.1155. [DOI] [PubMed] [Google Scholar]
  59. Brennan J, Karl J, Capel B. Divergent vascular mechanisms downstream of Sry establish the arterial system in the XY gonad. Dev Biol. 2002;244:418–28. doi: 10.1006/dbio.2002.0578. [DOI] [PubMed] [Google Scholar]
  60. Combes AN, Wilhelm D, Davidson T, Dejana E, Harley V, et al. Endothelial cell migration directs testis cord formation. Dev Biol. 2009;326:112–20. doi: 10.1016/j.ydbio.2008.10.040. [DOI] [PubMed] [Google Scholar]
  61. Cool J, Carmona FD, Szucsik JC, Capel B. Peritubular myoid cells are not the migrating population required for testis cord formation in the XY gonad. Sex Dev. 2008;2:128–33. doi: 10.1159/000143430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Tomizuka K, Horikoshi K, Kitada R, Sugawara Y, Iba Y, et al. R-spondin1 plays an essential role in ovarian development through positively regulating Wnt-4 signaling. Hum Mol Genet. 2008;17:1278–91. doi: 10.1093/hmg/ddn036. [DOI] [PubMed] [Google Scholar]
  63. Chassot AA, Ranc F, Gregoire EP, Roepers-Gajadien HL, Taketo MM, et al. Activation of beta-catenin signaling by Rspo1 controls differentiation of the mammalian ovary. Hum Mol Genet. 2008;17:1264–77. doi: 10.1093/hmg/ddn016. [DOI] [PubMed] [Google Scholar]
  64. Liu CF, Bingham N, Parker K, Yao HH. Sex-specific roles of beta-catenin in mouse gonadal development. Hum Mol Genet. 2009;18:405–17. doi: 10.1093/hmg/ddn362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Jeays-Ward K, Hoyle C, Brennan J, Dandonneau M, Alldus G, et al. Endothelial and steroidogenic cell migration are regulated by WNT4 in the developing mammalian gonad. Development. 2003;130:3663–70. doi: 10.1242/dev.00591. [DOI] [PubMed] [Google Scholar]
  66. Yao HH, Matzuk MM, Jorgez CJ, Menke DB, Page DC, et al. Follistatin operates downstream of Wnt4 in mammalian ovary organogenesis. Dev Dyn. 2004;230:210–5. doi: 10.1002/dvdy.20042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Tevosian SG, Manuylov NL. To beta or not to beta: canonical beta-catenin signaling pathway and ovarian development. Dev Dyn. 2008;237:3672–80. doi: 10.1002/dvdy.21784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Jordan BK, Shen JH, Olaso R, Ingraham HA, Vilain E. Wnt4 overexpression disrupts normal testicular vasculature and inhibits testosterone synthesis by repressing steroidogenic factor 1/beta-catenin synergy. Proc Natl Acad Sci USA. 2003;100:10866–71. doi: 10.1073/pnas.1834480100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Cool J, DeFalco TJ, Capel B. Vascular-mesenchymal cross-talk through Vegf and Pdgf drives organ patterning. Proc Natl Acad Sci USA. 2011;108:167–72. doi: 10.1073/pnas.1010299108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Combes AN, Lesieur E, Harley VR, Sinclair AH, Little MH, et al. Three-dimensional visualization of testis cord morphogenesis, a novel tubulogenic mechanism in development. Dev Dyn. 2009;238:1033–41. doi: 10.1002/dvdy.21925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Wu X, Wan S, Lee MM. Key factors in the regulation of fetal and postnatal Leydig cell development. J Cell Physiol. 2007;213:429–33. doi: 10.1002/jcp.21231. [DOI] [PubMed] [Google Scholar]
  72. Tapanainen J, Kellokumpu-Lehtinen P, Pelliniemi L, Huhtaniemi I. Age-related changes in endogenous steroids of human fetal testis during early and midpregnancy. J Clin Endocrinol Metab. 1981;52:98–102. doi: 10.1210/jcem-52-1-98. [DOI] [PubMed] [Google Scholar]
  73. Svechnikov K, Landreh L, Weisser J, Izzo G, Colón E, et al. Origin, development and regulation of human leydig cells. Horm Res Paediatr. 2010;73:93–101. doi: 10.1159/000277141. [DOI] [PubMed] [Google Scholar]
  74. Griswold SL, Behringer RR. Fetal leydig cell origin and development. Sex Dev. 2009;3:1–15. doi: 10.1159/000200077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Luo X, Ikeda Y, Parker KL. A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell. 1994;77:481–90. doi: 10.1016/0092-8674(94)90211-9. [DOI] [PubMed] [Google Scholar]
  76. Hatano O, Takayama K, Imai T, Waterman MR, Takakusu A, et al. Sex-dependent expression of a transcription factor, Ad4BP, regulating steroidogenic P-450 genes in the gonads during prenatal and postnatal rat development. Development. 1994;120:2787–97. doi: 10.1242/dev.120.10.2787. [DOI] [PubMed] [Google Scholar]
  77. Ikeda Y, Shen WH, Ingraham HA, Parker KL. Developmental expression of mouse steroidogenic factor-1, an essential regulator of the steroid hydroxylases. Mol Endocrinol. 1994;8:654–62. doi: 10.1210/mend.8.5.8058073. [DOI] [PubMed] [Google Scholar]
  78. Yao HH, Whoriskey W, Capel B. Desert Hedgehog/Patched 1 signaling specifies fetal Leydig cell fate in testis organogenesis. Genes Dev. 2002;16:1433–40. doi: 10.1101/gad.981202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, et al. A novel mutation of desert hedgehog in a patient with 46,XY partial gonadal dysgenesis accompanied by minifascicular neuropathy. Am J Hum Genet. 2000;67:1302–5. doi: 10.1016/s0002-9297(07)62958-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Canto P, Soderlund D, Reyes E, Mendez JP. Mutations in the desert hedgehog (DHH) gene in patients with 46,XY complete pure gonadal dysgenesis. J Clin Endocrinol Metab. 2004;89:4480–3. doi: 10.1210/jc.2004-0863. [DOI] [PubMed] [Google Scholar]
  81. Canto P, Vilchis F, Soderlund D, Reyes E, Mendez JP. A heterozygous mutation in the desert hedgehog gene in patients with mixed gonadal dysgenesis. Mol Hum Reprod. 2005;11:833–6. doi: 10.1093/molehr/gah216. [DOI] [PubMed] [Google Scholar]
  82. Barsoum I, Yao HH. Redundant and differential roles of transcription factors gli1 and gli2 in the development of mouse fetal Leydig cells. Biol Reprod. 2011;84:894–9. doi: 10.1095/biolreprod.110.088997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Barsoum IB, Bingham NC, Parker KL, Jorgensen JS, Yao HH. Activation of the Hedgehog pathway in the mouse fetal ovary leads to ectopic appearance of fetal Leydig cells and female pseudohermaphroditism. Dev Biol. 2009;329:96–103. doi: 10.1016/j.ydbio.2009.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Kitamura K, Yanazawa M, Sugiyama N, Miura H, Iizuka-Kogo A, et al. Mutation of ARX causes abnormal development of forebrain and testes in mice and X-linked lissencephaly with abnormal genitalia in humans. Nat Genet. 2002;32:359–69. doi: 10.1038/ng1009. [DOI] [PubMed] [Google Scholar]
  85. Brennan J, Tilmann C, Capel B. Pdgfr-alpha mediates testis cord organization and fetal Leydig cell development in the XY gonad. Genes Dev. 2003;17:800–10. doi: 10.1101/gad.1052503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Bowles J, Feng CW, Spiller C, Davidson TL, Jackson A, et al. FGF9 suppresses meiosis and promotes male germ cell fate in mice. Dev Cell. 2010;19:440–9. doi: 10.1016/j.devcel.2010.08.010. [DOI] [PubMed] [Google Scholar]
  87. Bowles J, Knight D, Smith C, Wilhelm D, Richman J, et al. Retinoid signaling determines germ cell fate in mice. Science. 2006;312:596–600. doi: 10.1126/science.1125691. [DOI] [PubMed] [Google Scholar]
  88. Koubova J, Menke DB, Zhou Q, Capel B, Griswold MD, et al. Retinoic acid regulates sex-specific timing of meiotic initiation in mice. Proc Natl Acad Sci USA. 2006;103:2474–9. doi: 10.1073/pnas.0510813103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. McLaren A, Southee D. Entry of mouse embryonic germ cells into meiosis. Dev Biol. 1997;187:107–13. doi: 10.1006/dbio.1997.8584. [DOI] [PubMed] [Google Scholar]
  90. Colvin JS, Feldman B, Nadeau JH, Goldfarb M, Ornitz DM. Genomic organization and embryonic expression of the mouse fibroblast growth factor 9 gene. Dev Dyn. 1999;216:72–88. doi: 10.1002/(SICI)1097-0177(199909)216:1<72::AID-DVDY9>3.0.CO;2-9. [DOI] [PubMed] [Google Scholar]
  91. MacLean G, Li H, Metzger D, Chambon P, Petkovich M. Apoptotic extinction of germ cells in testes of Cyp26b1 knockout mice. Endocrinology. 2007;148:4560–7. doi: 10.1210/en.2007-0492. [DOI] [PubMed] [Google Scholar]
  92. Kumar S, Chatzi C, Brade T, Cunningham TJ, Zhao X, et al. Sex-specific timing of meiotic initiation is regulated by Cyp26b1 independent of retinoic acid signalling. Nat Commun. 2011;2:151. doi: 10.1038/ncomms1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Childs AJ, Cowan G, Kinnell HL, Anderson RA, Saunders PT. Retinoic acid signalling and the control of meiotic entry in the human fetal gonad. PLoS ONE. 2011;6:e20249. doi: 10.1371/journal.pone.0020249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. le Bouffant R, Guerquin MJ, Duquenne C, Frydman N, Coffigny H, et al. Meiosis initiation in the human ovary requires intrinsic retinoic acid synthesis. Hum Reprod. 2010;25:2579–90. doi: 10.1093/humrep/deq195. [DOI] [PubMed] [Google Scholar]
  95. Cupp AS, Dufour JM, Kim G, Skinner MK, Kim KH. Action of retinoids on embryonic and early postnatal testis development. Endocrinology. 1999;140:2343–52. doi: 10.1210/endo.140.5.6720. [DOI] [PubMed] [Google Scholar]
  96. Spiller CM, Wilhelm D, Koopman P. Retinoblastoma 1 protein modulates XY germ cell entry into G1/G0 arrest during fetal development in mice. Biol Reprod. 2010;82:433–43. doi: 10.1095/biolreprod.109.078691. [DOI] [PubMed] [Google Scholar]
  97. Eicher EM, Washburn LL, Whitney JB, Morrow KE. Mus poschiavinus Y chromosome in the C57BL/6J murine genome causes sex reversal. Science. 1982;217:535–7. doi: 10.1126/science.7089579. [DOI] [PubMed] [Google Scholar]
  98. Albrecht KH, Young M, Washburn LL, Eicher EM. Sry expression level and protein isoform differences play a role in abnormal testis development in C57BL/6J mice carrying certain sry alleles. Genetics. 2003;164:277–88. doi: 10.1093/genetics/164.1.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Bullejos M, Koopman P. Delayed Sry and Sox9 expression in developing mouse gonads underlies B6-YDOM sex reversal. Dev Biol. 2005;278:473–81. doi: 10.1016/j.ydbio.2004.11.030. [DOI] [PubMed] [Google Scholar]
  100. Nagamine CM, Morohashi KI, Carlisle C, Chang DK. Sex reversal caused by Mus musculus domesticus Y chromosomes linked to variant expression of the testis-determining gene Sry. Dev Biol. 1999;216:182–94. doi: 10.1006/dbio.1999.9436. [DOI] [PubMed] [Google Scholar]
  101. Nef S, Schaad O, Stallings NR, Cederroth CR, Pitetti JL, et al. Gene expression during sex determination reveals a robust female genetic program at the onset of ovarian development. Dev Biol. 2005;287:361–77. doi: 10.1016/j.ydbio.2005.09.008. [DOI] [PubMed] [Google Scholar]
  102. Vainio S, Heikkila M, Kispert A, Chin N, McMahon AP. Female development in mammals is regulated by Wnt-4 signalling. Nature. 1999;397:405–9. doi: 10.1038/17068. [DOI] [PubMed] [Google Scholar]
  103. Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ. A WNT4 mutation associated with Müllerian-duct regression and virilization in a 46,XX woman. N Engl J Med. 2004;351:792–8. doi: 10.1056/NEJMoa040533. [DOI] [PubMed] [Google Scholar]
  104. Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, et al. The murine winged-helix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance. Development. 2004;131:933–42. doi: 10.1242/dev.00969. [DOI] [PubMed] [Google Scholar]
  105. Muroya K, Okuyama T, Goishi K, Ogiso Y, Fukuda S, et al. Sex-determining gene(s) on distal 9p: clinical and molecular studies in six cases. J Clin Endocrinol Metab. 2000;85:3094–100. doi: 10.1210/jcem.85.9.6771. [DOI] [PubMed] [Google Scholar]
  106. Crisponi L, Deiana M, Loi A, Chiappe F, Uda M, et al. The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome. Nat Genet. 2001;27:159–66. doi: 10.1038/84781. [DOI] [PubMed] [Google Scholar]
  107. Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, et al. Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development. Hum Mol Genet. 2004;13:1171–81. doi: 10.1093/hmg/ddh124. [DOI] [PubMed] [Google Scholar]
  108. Matson CK, Murphy MW, Sarver AL, Griswold MD, Bardwell VJ, et al. DMRT1 prevents female reprogramming in the postnatal mammalian testis. Nature. 2011;476:101–4. doi: 10.1038/nature10239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T, Sekido R, et al. Somatic sex reprogramming of adult ovaries to testes by FOXL2 ablation. Cell. 2009;139:1130–42. doi: 10.1016/j.cell.2009.11.021. [DOI] [PubMed] [Google Scholar]
  110. Sadovsky Y, Crawford PA, Woodson KG, Polish JA, Clements MA, et al. Mice deficient in the orphan receptor steroidogenic factor 1 lack adrenal glands and gonads but express P450 side-chain-cleavage enzyme in the placenta and have normal embryonic serum levels of corticosteroids. Proc Natl Acad Sci USA. 1995;92:10939–43. doi: 10.1073/pnas.92.24.10939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Fatchiyah, Zubair M, Shima Y, Oka S, Ishihara S, et al. Differential gene dosage effects of Ad4BP/SF-1 on target tissue development. Biochem Biophys Res Commun. 2006;341:1036–45. doi: 10.1016/j.bbrc.2006.01.058. [DOI] [PubMed] [Google Scholar]
  112. Bland ML, Fowkes RC, Ingraham HA. Differential requirement for steroidogenic factor-1 gene dosage in adrenal development versus endocrine function. Mol Endocrinol. 2004;18:941–52. doi: 10.1210/me.2003-0333. [DOI] [PubMed] [Google Scholar]
  113. Hasegawa T, Fukami M, Sato N, Katsumata N, Sasaki G, et al. Testicular dysgenesis without adrenal insufficiency in a 46,XY patient with a heterozygous inactive mutation of steroidogenic factor-1. J Clin Endocrinol Metab. 2004;89:5930–5. doi: 10.1210/jc.2004-0935. [DOI] [PubMed] [Google Scholar]
  114. Mallet D, Bretones P, Michel-Calemard L, Dijoud F, David M, et al. Gonadal dysgenesis without adrenal insufficiency in a 46,XY patient heterozygous for the nonsense C16X mutation: a case of SF1 haploinsufficiency. J Clin Endocrinol Metab. 2004;89:4829–32. doi: 10.1210/jc.2004-0670. [DOI] [PubMed] [Google Scholar]
  115. Swain A, Zanaria E, Hacker A, Lovell-Badge R, Camerino G. Mouse Dax1 expression is consistent with a role in sex determination as well as in adrenal and hypothalamus function. Nat Genet. 1996;12:404–9. doi: 10.1038/ng0496-404. [DOI] [PubMed] [Google Scholar]
  116. Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC, et al. A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet. 1994;7:497–501. doi: 10.1038/ng0894-497. [DOI] [PubMed] [Google Scholar]
  117. Muscatelli F, Strom TM, Walker AP, Zanaria E, Recan D, et al. Mutations in the DAX-1 gene give rise to both X-linked adrenal hypoplasia congenita and hypogonadotropic hypogonadism. Nature. 1994;372:672–6. doi: 10.1038/372672a0. [DOI] [PubMed] [Google Scholar]
  118. Zanaria E, Muscatelli F, Bardoni B, Strom TM, Guioli S, et al. An unusual member of the nuclear hormone receptor superfamily responsible for X-linked adrenal hypoplasia congenita. Nature. 1994;372:635–41. doi: 10.1038/372635a0. [DOI] [PubMed] [Google Scholar]
  119. Bouma GJ, Washburn LL, Albrecht KH, Eicher EM. Correct dosage of Fog2 and Gata4 transcription factors is critical for fetal testis development in mice. Proc Natl Acad Sci USA. 2007;104:14994–9. doi: 10.1073/pnas.0701677104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Eicher EM, et al. Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development. 2002;129:4627–34. doi: 10.1242/dev.129.19.4627. [DOI] [PubMed] [Google Scholar]
  121. Tevosian SG, Deconinck AE, Cantor AB, Rieff HI, Fujiwara Y, et al. FOG-2: A novel GATA-family cofactor related to multitype zinc-finger proteins Friend of GATA-1 and U-shaped. Proc Natl Acad Sci USA. 1999;96:950–5. doi: 10.1073/pnas.96.3.950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Park SY, Lee EJ, Emge D, Jahn CL, Jameson JL. A phenotypic spectrum of sexual development in Dax1 (Nr0b1)-deficient mice: consequence of the C57BL/6J strain on sex determination. Biol Reprod. 2008;79:1038–45. doi: 10.1095/biolreprod.108.069492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Meeks JJ, Crawford SE, Russell TA, Morohashi K, Weiss J, et al. Dax1 regulates testis cord organization during gonadal differentiation. Development. 2003;130:1029–36. doi: 10.1242/dev.00316. [DOI] [PubMed] [Google Scholar]
  124. Meeks JJ, Weiss J, Jameson JL. Dax1 is required for testis determination. Nat Genet. 2003;34:32–3. doi: 10.1038/ng1141. [DOI] [PubMed] [Google Scholar]
  125. Bouma GJ, Albrecht KH, Washburn LL, Recknagel AK, Churchill GA, et al. Gonadal sex reversal in mutant Dax1 XY mice: a failure to upregulate Sox9 in pre-Sertoli cells. Development. 2005;132:3045–54. doi: 10.1242/dev.01890. [DOI] [PubMed] [Google Scholar]
  126. Ludbrook LM, Harley VR. Sex determination: a ‘window' of DAX1 activity. Trends Endocrinol Metab. 2004;15:116–21. doi: 10.1016/j.tem.2004.02.002. [DOI] [PubMed] [Google Scholar]