Quick viewing(Text Mode)

Genetic and Epigenetic Effects in Sex Determination Sezgin Ozgur Gunes1,2, Asli Metin Mahmutoglu1, and Ashok Agarwal*3

Genetic and Epigenetic Effects in Sex Determination Sezgin Ozgur Gunes1,2, Asli Metin Mahmutoglu1, and Ashok Agarwal*3

Genetic and Epigenetic Effects in Sex Determination Sezgin Ozgur Gunes1,2, Asli Metin Mahmutoglu1, and Ashok Agarwal*3

Sex determination is a complex and dynamic process with multiple genetic cascade are not completely understood. This review aims at discussing and environmental causes, in which germ and somatic cells receive various current data on the genetic effects via genes and epigenetic mechanisms that sex-specific features. During the fifth week of fetal life, the bipotential affect the regulation of sex determination. embryonic gonad starts to develop in humans. In the bipotential gonadal tissue, certain cell groups start to differentiate to form the or testes. Birth Defects Research (Part C) 108:321–336, 2016. Despite considerable efforts and advances in identifying the mechanisms VC 2016 Wiley Periodicals, Inc. playing a role in sex determination and differentiation, the underlying mechanisms of the exact functions of many genes, gene–gene interactions, Key words: sex determination; SRY; SOXE; NR5A1; GATA4; WT1; epigenetics and epigenetic modifications that are involved in different stages of this

Introduction formation via inducing a different set of genes (Sekido and Sex determination is a biological process determining the Lovell-Badge, 2008; Rigby and Kulathinal, 2015). development of the primordial gonad into male (testes) or Animal experiments and genetic analyses in patients () gonads (Herpin and Schartl, 2011). During with developmental sex disorders (DSDs) have demonstrat- the sex determination cascade, the initial event is the forma- ed that many genes and pathways, such as GATA4, SOX9, tion of the gonadal primordium, also known as gonadal or NR5A1, FOG2, Hedgehog, and the Map Kinase signaling path- genital ridge (Ronfani and Bianchi, 2004). The gonadal pri- way, play critical roles in the sex determination process mordium derives from the mesonephros that is one of the (Cotinot et al., 2002). Recent studies have proposed that epi- tubular nephric structures of the urogenital ridges in mam- genetic mechanisms are also involved in the regulation of mals (Lucas-Herald and Bashamboo, 2014), and is coated by sex determination (Kuroki et al., 2013; Mulvey et al., 2014; coelomic epithelium (Wilhelm et al., 2013). Epithelial cells Skinner et al., 2015). In this review, current and past litera- derived from the proliferation of the epithelium of the ture on genetic effects on the regulation of sex determina- gonadal primordium enter the mesenchyme and result in tion, with special emphasis on male sex determination, is the formation of primitive sex cords. These sex cords lose discussed. In addition, the contribution of epigenetic mecha- nisms to the process of sex determination is reviewed. connections with the epithelial surface, share the same morphology in fetuses with XX or XY sex chromosomes, and SRY are called bipotential gonads (Matzuk and Lamb, 2008; Sex Determining Region Y (SRY) (OMIM *480000) is a single Sadler, 2011). exon gene and encodes a DNA binding protein, a member of A variety of genes including WT1, FOG2, and NR5A1 the high mobility group (HMG)-box family. The SRY gene enco- are known to play a role in the bipotential gonad forma- des a and is located on the position p11.3 tion (Barrionuevo et al., 2012). The differentiation of bipo- of the Y-chromosome (Premi et al., 2006). This transcription tential gonads along the testis- or ovary-specific pathway factor is called the testis-determining factor (TDF), induces is induced by SRY gene expression (Wijchers and Festen- male sex determination, and has been proposed to play a piv- stein, 2011). The expression of the SRY gene gives rise to otal role in sex determination in males (Page et al., 1987). The the activation of downstream genes involved in testis for- SRY transcript has many transcription starting sites, with the mation by inducing Sertoli-cell differentiation, whereas the size of the transcript being approximately 900 nucleotides dysfunction or absence of SRY expression results in ovary (Clepet et al., 1993). The SRY protein is constituted of N- terminal, central, and C-terminal domains containing the HMG box which is an evolutionary conserved DNA binding 1 Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis and bending domain spanning over 79 amino acids. The HMG University, Samsun, Turkey 2Department of Multidisciplinary Molecular Medicine, Ondokuz Mayis box motif provides binding to a specific DNA sequence via University, Health Science Institute, Samsun, Turkey interaction with the minor groove and causes bending of the 3American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, DNA helix up to a 908 angle. This bending enables the forma- Ohio tion of an active transcriptional complex on the DNA. Protein–

*Correspondence to: Ashok Agarwal, Lerner College of Medicine, Andrology protein interactions and post-translational modifications also Center and the American Center for Reproductive Medicine, Cleveland take place in the HMG domain. Thus, since most of the muta- Clinic, Mail Code X-11, 10681 Carnegie Avenue, Cleveland, OH 44195. tions occur de novo in the DNA sequence encoding this E-mail: [email protected] domain, and these mutations cause to abnormalities in DNA Published online in Wiley Online Library (wileyonlinelibrary.com). Doi: 10.1002/bdrc. binding and sex reversal, the HMG box domain has a very crit- 21146 ical role (Helszer et al., 2013). HMG box domain mutations

VC 2016 Wiley Periodicals, Inc. 322 MOLECULAR MECHANISMS IN SEX DETERMINATION

also change nuclear importation and are involved in the pancreas specification, gliogenesis, and neural crest devel- minority of human sex reversal cases (Sim et al., 2008). opment (Huang et al., 2015). SOXE proteins contain the Alterations in the DNA sequence of the SRY gene can transcription activation domain, as well as the DNA- result in , which is one of the DSDs binding HMG domain, similar to all other SOX proteins. (Schlessinger et al., 2010). DSDs are congenital abnormities Moreover, the DNA-dependent dimerization domain is a with numerical or structural chromosomal aberrations, unique structure of SOXE proteins (Barrionuevo and gonadal, or anatomic sex development abnormalities (Ohne- Scherer, 2010). In mammals, SOXE genes are involved in sorg et al., 2014). 46,XX, 46,XY and sex chromosome DSDs sex determination (She and Yang, in press) via enhancing are categories of developmental sex disorders, according to the activities of testis-specific enhancer (TES) of Sox9 core the Chicago Consensus (Hughes et al., 2006). The aneuploi- element (TESCO) and anti-Mullerian Hormone (Amh)pro- dies of X- and/or Y-chromosomes cause sex chromosome moter (Otake and Kuroiwa, 2016). DSDs, including (TS), Klinefelter syn- In 46,XY embryos, gonadal differentiation is initiated by drome (KS), and 45,X/46,XY mosaicism. Children with KS or SRY expression between 41 and 44 days postovulation TS have no genital differences in their early lives; whereas, (d.p.o.)/Carnegie stages 18 (CS18). Once SRY expression children with the mosaic 45,X/46,XY have ambig- peaks at 44 d.p.o./CS18, a period of time when sex cords are uous genitalia in their newborn period. Although 46,XX first seen, SOX9 gene activation is initiated in the bipotential DSDs and 46,XY DSDs result from different conditions, high gonad and triggers Sertoli cell differentiation. The expres- levels of exposure during the fetal period and sion of the SRY gene continues up to 18-weeks gestation incomplete intrauterine , respectively, they all (Hanley et al., 2000; Eggers et al., 2014). SOX9 gene expres- cause the generation of external genitalia improper to sex sion is promoted by not only SRY expression, but also chromosome constitution (Achermann et al., 2015). enhancers located in different regions upstream of SOX9 To date, more than 50 mutations of the SRY gene have gene, depending on tissue type and time (Lybaek et al., been associated with sex reversal (Andonova et al., 2015). 2014). TESCO is one of the regulatory elements of the SOX9 These mutations are summarized in Table 1. Most of the gene and engages an important role as an enhancer in sex mutations of SRY gene associated with gonadal dysgenesis determination of the mammalian organisms (Kimura et al., have been detected in the HMG box. Helszer et al. (2013) 2014). DNA lesions leading to changes in the upstream regu- detected a novel missense mutation (c.341A>G) causing the lation of SOX9 have been suggested to be related to SRY neg- substitution of asparagine by aspartic acid at codon 65 ative XX DSDs (Kojima et al., 2008). Benko et al. (2011) have (p.N65D) in the HMG box of SRY gene. The binding ability of reported duplications and deletions in the SOX9 regulatory the N65D variant of the SRY was analyzed by gel retardation region in patients with isolated 46,XX and 46,XY DSDs, analysis. These authors suggested that the c.341A>Gmuta- respectively. Although duplication in the upstream region of tion may also contribute to the pathogenesis of 46,XY gonadal SOX9 is known to cause SRY2 XX DSDs (Hyon et al., 2015), dysgenesis (Helszer et al., 2013). Although M64I is a classical polymorphisms of SOX9 do not result in SRY2 XX DSDs (Xia sex reversal mutation at position 9 in the HMG box, it has no et al., 2015). Recently, a study has proposed that the regula- adverse effect on DNA binding (McElreavey et al., 1992). tory regions of the SOX9 gene span two different regions, XY Another mutation with no effect on DNA binding activi- and XX sex reversal regions. Therefore, deletions or duplica- ty is the F108S mutation, a familial mutation in the SRY tions cause distinct forms of DSDs (Kim et al., 2015). gene (J€ager et al., 1992). More recently, the F108S mutation SOX9 has been found to be associated with elevated polyubiquiti- SOX9 (OMIM *608160) is an autosomal gene located on nation. This process is a modification correlated with an 17q24.3, and is a primary downstream target of SRY (Foster increase in proteosomal degradation. Polyubiquitination of et al., 1994). SOX9 protein has very critical and diverse func- mutant SRY has been suggested to lead to an increase in tions in embryonic development (Katoh-Fukui et al., 2015), proteosomal degradation, resulting in a reduction of intra- such as sex determination by initiating male-specific sex cellular level of SRY. The alteration of intracellular SRY lev- determination in the default of Sry in Sertoli cells (Kent els caused a reduction in transcriptional activation of Sox9, et al., 1996; Vidal et al., 2001; Chaboissier et al., 2004), carti- which can obstruct normal gonadogenesis (Gerhardt, 2016; lage formation (Bi et al., 1999), and neural crest (Spokony Racca et al., 2016). These mutations also demonstrate that et al., 2002; Cheung and Briscoe, 2003), skeletal (Foster SRY is a cornerstone in sex determination. However, apart et al., 1994, Wagner et al., 1994), brain (Pompolo and Harley, from SRY, other genes are also essential for gonadal devel- 2001), and craniofacial development (Lee and Saint-Jeannet, opment in males, such as SOX9, NR5A1,andGATA4 (Fig. 1). 2011). SOX9 heterozygous mutations are involved in Campo- SOXE GENES melic Dysplasia identified with defects in bone morphology SRY-related HMG box (SOX)8, SOX9, and SOX10 proteins (Gordon et al., 2014) and sex reversal (Berta et al., 1990). are SOXE transcription factors which are implicated in var- Ortego et al. (2015) colleagues have shown that overexpres- ious developmental processes including sex determination, sion of Sox9 promotes testis determination in male mice IT EET EERH(ATC 0:2–3 (2016) 108:321–336 C) (PART RESEARCH DEFECTS BIRTH

TABLE 1. Studies Reporting Mutations of SRY Gene

Type of Amino acid Location of mutation change mutation in SRY Effects of mutation Transmission Phenotype Method Reference

Missense Gly ! Arg HMG Box DNA binding De novo Gonadal Dysgenesis SSCP assay and DNA Hawkins et al. (1992a) activity may change Sequencing

Nonsense NA HMG Box DNA binding De novo Gonadal Dysgenesis SSCP assay and DNA Hawkins et al. (1992a)

activity may change Sequencing

Missense Phe ! Ser HMG Box No differences in Familial Gonadal Dysgenesis DNA Sequencing Jager€ et al. (1992) DNA binding

activity

Nonsense Gln ! Stop ORF DNA binding De novo Pure Gonadal Dysgenesis Asymetric PCR and DGGE McElreavey et al. (1992) activity may change

Deletion – 5’ to the HMG Box NA De novo Pure Gonadal Dysgenesis Southern Blot Analysis McElreavey et al. (1992)

Missense Ile ! Thr HMG Box NA De novo Pure Gonadal Dysgenesis DGGE and DNA Sequencing McElreavey et al. (1992)

Nonsense Tyr ! Stop HMG Box NA De novo Pure Gonadal Dysgenesis DGGE and DNA Sequencing McElreavey et al. (1992)

Nonsense Gln ! Stop HMG Box NA De novo Pure Gonadal Dysgenesis DGGE and DNA Sequencing McElreavey et al. (1992) Missense Met ! Ile HMG Box NA De novo Pure Gonadal Dysgenesis DGGE and DNA Sequencing McElreavey et al. (1992)

Missense Val ! Leu ORF NA Familial Gonadal Dysgenesis DNA Sequencing Vilain et al. () 46, XY Male

Nonsense Lys ! Stop ORF NA De novo Gonadal Dysgenesis DNA Sequencing Muller€ et al. (1992)

Missense Ile ! Met ORF NA Familial Complete Gonadal SSCP assay and Hawkins et al. (1992b) Dysgenesis DNA Sequencing

Missense Leu ! Ile ORF NA NA Complete Gonadal SSCP assay and Hawkins et al. (1992b) Dysgenesis DNA Sequencing

Deletion – ORF NA De novo Complete Gonadal Dysgenesis SSCP assay and Hawkins et al. (1992b)

DNA Sequencing

Missense Arg ! Gly HMG Box NA De novo Pure Gonadal Dysgenesis DNA Sequencing Affara et al. (1993)

Missense Met ! Trp HMG Box NA De novo Pure Gonadal Dysgenesis DNA Sequencing Affara et al. (1993)

Missense Arg ! Trp HMG Box NA NA Pure Gonadal Dysgenesis DNA Sequencing Affara et al. (1993)

Nonsense Gln ! Stop HMG Box NA NA Pure Gonadal Dysgenesis DNA Sequencing Affara et al. (1993)

Silent Leu ! His SRY NA Post Zigotic DNA Sequencing Braun et al. (1993) Deletion – Outside of TDF NA Induced (Mice) Gonadal Dysgenesis Southern Blot Analysis Capel et al. (1993) 323 324

TABLE 1. Continued

Type of Amino acid Location of mutation change mutation in SRY Effects of mutation Transmission Phenotype Method Reference

Missense Ala ! Thr HMG Box No or reduced De novo Gonadal Dysgenesis DNA Sequencing Zeng et al. (1993) DNA binding

activity

Nonsense Trp ! Stop HMG Box NA De novo Gonadal Dysgenesis SSCP assay and DNA Iida et al. (1994) Sequencing

Nonsense Leu ! Stop 30 Non-HMG Box NA Familial Gonadal Dysgenesis SSCP assay and DNA Tajima et al. (1994) Sequencing

Missense Ser ! Gly HMG Box Reduced DNA De novo Gonadal Dysgenesis SSCP Assay Schmitt-Ney binding activity et al. (1995)

Missense Pro ! Leu HMG Box Reduced DNA Familial Gonadal Dysgenesis SSCP Assay Schmitt-Ney binding activity et al. (1995)

Interstitial –30 to SRY NA De novo Partial Gonadal Southern Blot Analysis McElreavey

Deletion Dysgenesis et al. (1996)

Nonsense Gln ! Stop ORF Incomplete DNA Familial Gonadal Dysgenesis DNA Sequencing Bilbao et al. (1996) binding

Nonsense Trp ! Stop ORF NA De novo Complete Gonadal Dysgenesis FAMA Veitia et al. (1997)

Missense Arg ! Trp ORF NA De novo Complete Gonadal Dysgenesis DNA Sequencing Veitia et al. (1997)

Missense Arn ! Asn HMG Box NA De novo Gonadal Dysgenesis SSCP Assay and DNA Battiloro et al. (1997) Sequencing OEUA EHNSSI E DETERMINATION SEX IN MECHANISMS MOLECULAR Point – 50 to SRY NA NA Pure Gonadal Dysgenesis SSCP Assay and DNA Poulat et al. (1998)

Sequencing

Nonsense Lys ! Stop 50 to HMG Box NA De novo Gonadal Dysgenesis SSCP Assay and DNA Scherer et al. (1998) Sequencing

Missense Met ! Arg HMG Reduced DNA De novo Gonadal Dysgenesis SSCP Assay and DNA Scherer et al. (1998) binding activity Sequencing

Missense Phe ! Val HMG Reduced DNA De novo Gonadal Dysgenesis SSCP Assay and DNA Scherer et al. (1998) binding activity Sequencing

Missense Ser ! Asn 50 Non-HMG Box DNA binding Familial Partial Gonadal Dysgenesis DNA Sequencing Domenice activity may change et al. (1998)

Nonsense Tyr ! Stop De novo 45,X/47,XYY Mosaicism DNA Sequencing Takagi et al. (1999) IT EET EERH(ATC 0:2–3 (2016) 108:321–336 C) (PART RESEARCH DEFECTS BIRTH

TABLE 1. Continued

Type of Amino acid Location of mutation change mutation in SRY Effects of mutation Transmission Phenotype Method Reference

Upstream of Lack of DNA

HMG Box binding motif

Missense Ser!Asn 50 Non-HMG Box NA De novo Ullrich-Turner Syndrome DNA Sequencing Canto et al. (2000)

Missense Gly ! Gln HMG Box NA De novo Complete Gonadal DNA Sequencing Schaffler et al. (2000) Dysgenesis with Yolk-Sac Tumor

Missense Arg ! Ile Non-HMG Box NA Familial Partial Gonadal DNA Sequencing Assumpcao et al. (2002) Dysgenesis

46, XY Male

Missense Asn ! His HMG Box Altered DNA De novo Pure Gonadal DNA Sequencing Assumpcao et al. (2002) binding activity Dysgenesis

Missense Arg ! Asn HMG Box No DNA binding De novo Complete Gonadal – Mitchell and Harley (2002) activity Dysgenesis

Missense Leu ! Pro HMG Box No DNA binding NA Complete Gonadal – Mitchell and Harley (2002) activity Dysgenesis

Missense Phe ! Val HMG Box Disrupted DNA De novo Complete Gonadal – Mitchell and Harley (2002) binding activity Dysgenesis

Missense Met ! Arg HMG Box Reduced DNA De novo Complete Gonadal – Mitchell and Harley (2002) binding activity (6 Dysgenesis

Fold)

Missense Arg ! Pro HMG Box Reduced DNA NA Complete Gonadal – Mitchell and Harley (2002) binding activity (4 Dysgenesis

Fold)

Missense Ala ! Thr HMG Box Reduced DNA De novo Complete Gonadal – Mitchell and Harley (2002) binding activity (3 Dysgenesis

Fold)

Missense Met ! Thr HMG Box Reduced DNA NA Complete Gonadal – Mitchell and Harley (2002) binding activity (1.7 Dysgenesis

Fold) 325 326

TABLE 1. Continued

Type of Amino acid Location of mutation change mutation in SRY Effects of mutation Transmission Phenotype Method Reference

Missense Ser ! Asn Outside the Minimal effect on Familial Partial Gonadal – Mitchell and Harley (2002) HMG Box DNA binding Dysgenesis

activity

Missense Tyr ! Phe HMG Box NA Familial Gonadal Dysgenesis DNA Sequencing Jordan et al. (2002) 46, XY Male

Missense Ile ! Met HMG Box DNA binding Familial True Hermophroditism DNA Sequencing Maier et al. (2003) activity may change

Missense Arg ! Trp NLS Reduction in MPB De novo Gonadal Dysgenesis – Harley et al. (2003) binding

Missense Arg ! Gly NLS Reduction in MPB De novo Gonadal Dysgenesis – Harley et al. (2003) binding

Missense Arg ! Asn NLS Normal MPB De novo Gonadal Dysgenesis – Harley et al. (2003) binding

Missense Arg ! Pro NLS NA NA Gonadal Dysgenesis – Harley et al. (2003)

Missense Glu ! Arg ORF NA De novo Gonadal Dysgenesis SSCP assay and DNA Shahid et al. (2004) Sequencing

Missense Glu ! Arg Upstream of NA De novo Gonadal Dysgenesis SSCP assay and DNA Shahid et al. (2004) HMG Box Sequencing

Missense Ser ! Cys Downstrem of NA De novo Gonadal Dysgenesis SSCP assay and DNA Shahid et al. (2004) OEUA EHNSSI E DETERMINATION SEX IN MECHANISMS MOLECULAR HMG Box Sequencing

Nonsense Asn ! Stop HMG Box DNA binding De novo 45,X/46,XY Mosaicism SSCP assay and DNA Shahid et al. (2005) activity may change Sequencing

Nonsense Leu ! Stop Downstrem of DNA binding De novo 45,X/46,XY Mosaicism SSCP assay and DNA Shahid et al. (2005) HMG Box activity may change Sequencing

Missense Gln ! His HMG Box DNA binding De novo 45,X/46,XY Mosaicism SSCP assay and DNA Shahid et al. (2005) activity may change Sequencing

Missense Trp ! Leu NLS Reduction in De novo Gonadal Dysgenesis DNA Sequencing Hersmus et al. (2009) nuclear import with Bilateral

Gonadoblastoma BIRTH DEFECTS RESEARCH (PART C) 108:321–336 (2016) 327

with deleted Sry gene, but also leads to aberrant testicular development, abnormalities in seminiferous epithelium with normal sperm count, motility, morphology, and fertilization capacity (Ortega et al., 2015). In the case of the absence of Sox9 in Sertoli cells, Sox8,orSox10 could be functional (Eggers et al., 2014).

SOX8 et al. (2012) SOX8 (OMIM *605923), 5.6 kb in size, is located on the petit arm of chromosome 16 and codes the SOXE tran- scription factor that is expressed in branchial arches, pan- creas, brain, heart, limbs, and testis. SOX8 transcription encoded by SOX8 gene is quite similar to other SOXE pro- teins, in terms of the sequence similarity within and out- side of the HMG domain. SOX8 deletion is involved in a- thalassemia/mental retardation syndrome (ATR-16) (Pfei- DNA SequencingDNA Sequencing Paliwal et al. (2011) Paliwal et al. (2011) DNA Sequencing Filges et al. (2011) DNA Sequencing Helszer et al. (2013) DNA Sequencingfer Andonova et al. et (2015) al., 2000; Schepers et al., 2003). Sox8 is one of the definitive markers of Sertoli cells and consolidates Sox9 function in gonad determination. Following the Sox9 acti- vation, Sox8 expression may induce differentiation in a Sertoli cell-specific manner (Chaboissier et al., 2004). In addition, a study conducted with Sox8 knockout mice has shown that Sox8 is vital for male fertility, since the absence of this protein causes an abnormality in the regu- lation of spermatogenesis in an age-dependent manner and progressive degeneration of the seminiferous epitheli- Dysgenesis Dysgenesis Dysgenesis Complete Gonadal Dysgenesis Complete Gonadal Dysgenesis um (O’Bryan et al., 2008). Hence, expression of Sox8 and Sox9 provides for proper testis cord formation (Barrio- nuevo et al., 2009) through maintaining the integrity of the basal lamina of the testis during testis development (Georg et al., 2012). A more recent study has proposed FamilialFamilial Pure Gonadal Pure Gonadal Familial Gonadal Dysgenesis RFLP Stoppa-Vaucher Familial Complete Gonadal De novo De novo that Sox8 and Sox9 provide constant expression of Dmrt1 in adult testis, which is essential to maintain functionality and integrity of seminiferous tubules, suggesting a new function for these genes. Sox8 and Sox9 are antiapoptotic factors in adult Sertoli cells (Barrionuevo et al., 2016).

SOX10

Effects of mutation Transmission Phenotype MethodSOX10 (OMIM Reference *602229), spanning five exons, is one of the activity may change activity may change binding activity binding activity binding activity SOXE genes (She and Yang, in press). Sox10 and SOX10 genes have been cloned from mouse and human, respectively, and characterized in 1998 (Pusch et al., 1998). Furthermore, SRY SOX10 protein shares 59% of its amino acid sequence identi- ty with SOX9 protein (Pusch et al., 1998). SOX10 has a role in

Location of oligodendrocyte development and differentiation of periph- mutation in eral glial cells, as well as sex determination and differentia- tion (Britsch et al., 2001; Cory et al., 2007; Chaoui et al., 2015). Leu HMG Box NA Asp HMG Box Reduced DNA Ser HMG Box Reduced DNA Leu HMG Box DNA binding Ala HMG Box Reduced DNA Stop HMG Box DNA binding SOX10 mutations result in Waardenburg-Hirschsprung ! ! ! ! ! !

change disease and Kallmann syndrome (MIM no. 147950) (Kuhl- Amino acid brodt et al., 1998; Pingault et al., 1998; Chaoui et al., 2015). Sox10 is expressed in pre-Sertoli cells in the male genital Continued ridge in mice. However, no abnormality of testis develop- ment has been reported in knockout mice with lost Sox10 Type of mutation Nonsense Tyr Missense Glu Missense Leu NA, not applicable. Missense Asn Missense Glu Missense Phe

TABLE 1. function (Cory et al., 2007). Polanco et al. (2010) have 328 MOLECULAR MECHANISMS IN SEX DETERMINATION

FIGURE 1. Gonadal development in the male. Multiple genes and pathways required for differentiation of bipotential gonad into testis, including NR5A1, GATA4, and WT1 (1KTS isoform), upregulate the master switch gene, SRY. Activation of SRY gene in the bipotential gonad initiates SOX9 expression, and then SOX9 promotes AMH expression in conjunction with NR5A1. FOG2 and GATA4 are also essential for AMH expression and testis development.

FIGURE 2. Epigenetic mechanisms in sex determination. The figure identifies main epigenetic mechanisms involved in sex determination process. Activation of genes involved in this process is regulated by histone modifications and DNA methylation. Gonadal functions are also organized by noncoding RNAs. shown that one of the reasons for sex reversal syndrome is possess DNA sequence similarity in the HMG box domain Sox10 overexpression. Expression levels of the Sox10 gene (Stevanovlc´ et al., 1993). Therefore, the SRY gene has been have been shown to be associated with various degrees of suggested to originate from SOX3 (Foster and Graves, sex reversal in transgenic mice (Polanco et al., 2010). 1994). Sox3 is expressed in the brain and developing gonads, and plays critical roles in neuronal development SOX3 and gonadal function. Weiss et al. (2003) have shown that SOX3 (OMIM *313430) gene has a single exon (Rizzoti Sox3 deletion has no adverse effect on sex determination et al., 2004), belongs to the SOX gene family, and has been in the male or female. However, Sox3 is essential for game- cloned and mapped on the long arm of the togenesis in both sexes (Collignon et al., 1996; Bylund in the human. Human and mouse SOX3 genes show high et al., 2003; Weiss et al., 2003). Sox3 runs with steroido- DNA sequence similarity. Moreover, SOX3 and SRY also genic factor 1(Sf1) to transactivate TESCO, similar to Sry. BIRTH DEFECTS RESEARCH (PART C) 108:321–336 (2016) 329

Inactivation or loss-of-function SOX3 mutations have not hormone (AMH) in Sertoli cells, also known as Mullerian been associated with sex determination either in mice or inhibiting substance, causing the regression of the parame- humans, however, are related with growth hormone defi- sonephric (Mullerian) ducts; and therefore, the and ciency or mental retardation. Conversely, chromosomal uterine tube, the upper part of cannot be differenti- rearrangements or gain-of-function mutations may lead to ated from these ducts in the sixth week of gestation. After 46,XX male sex reversal (Sutton et al., 2011; Gunes et al., this time, while expression of SF1 increases in Leydig cells, 2013). Two recent publications, demonstrated that dupli- expression of SF1 decreases in Sertoli cells. Apart from cation of the chromosomal region incorporating the SOX3 SF1 and SOX9, GATA4, and WT1 also participate in the reg- gene led to the sex reversal syndrome in one case (Moa- ulation of AMH secretion (Shen et al., 1994; De Santa Bar- lem et al., 2012), and an 774-kb insertional translocation bara et al., 2000). Furthermore, in Leydig cells, SF1 from chromosome 1 to the distal SOX3 gene led to altera- induces the synthesis of testicular leading to tions in SOX3 expression in a patient with 46,XX male sex differentiation of the mesonephric (Wolffian) ducts into reversal, suggesting that SOX3 can be a switch regulatory male urogenital structures (Parker et al., 1999). gene for promoting sex determination in humans (Haines NR5A1 gene loss-of-function mutations have been con- et al., 2015). nected with various genotypes and phenotypes of DSDs, such as 46,XX ovo-testicular DSD (Baetens et al., in press), NR5A1 46,XY DSD with adrenal failure (Achermann et al., 1999) SF1 is one of the transcription factors identified and and hypospadias (Tantawy et al., 2014), and also 46,XX involved in the modulation of sex determination and ste- DSD with primary (Philibert et al., 2010). In roidogenesis pathways, and development of the reproduc- tive axis and spleen (Oba et al., 1996; Achermann et al., children and adults with a 46,XX karyotype from unrelated 2002; Zangen et al., 2014). SF1 is encoded by the nuclear families, testis development first has been reported to be receptor subfamily 5 group A1 (NR5A1) (OMIM *184757), connected with a missense mutation in the DNA binding also initially known as FTZ-F1 and AD4BP, and has been region of NR5A1 (p.Arg92Trp). The mutation causes non- mapped to chromosome 9q33 by using fluorescence in syndromic 46,XX testicular or ovo-testicular DSD in situ hybridization in the early 1990s (Taketo et al., 1995). humans (Bashamboo et al., 2006). Recently, a novel mater- Its genomic sequence was identified by cloning of human nally inherited NR5A1 mutation (c.195G > A) has also been cDNA (Wong et al., 1996). The NR5A1 gene spans seven found in three siblings with 46,XY DSDs (Fabbri et al., exons with a noncoding exon, and is about 30 kb in length 2014). (Oba et al., 1996). Exon 2 and 3 are the first 2 coding GATA4 AND WT1 exons of NR5A1 and encode two zinc finger domains GATA4 and WT1 are the two transcription factors products which are conserved in rat, mouse, cow, and human, and of having a role in the sex determination process similar necessary for DNA binding. Mutation in these exons to multiple other genes (Table 2). In the sex determination caused NR5A1 haploinsufficiency in a presenting female and differentiation pathways, transcriptions of certain patient with 46,XY gonadal dysgenesis who had external genes have been controlled by the cooperation of GATA4 genitalia with , absence of the uterus, and a and WT1 (Miyamoto et al., 2008). Wilms’ tumor (WT1) mild form of dysgenetic testes (Barbaro et al., 2011). SF1 expression starts with the sex cord formation and Sertoli binds to the promoters of more than 20 genes, has func- cell differentiation during the early stages of the human tions in male , steroidogenesis, and embryo (from 6.5 to 8 weeks) and continues with a high reproduction with different roles as a monomer unlike level of expression up to the nineth week of gestation. other transcriptional factors and regulates their transcrip- After sex cord formation, GATA4 and WT1 expression is tion (Achermann et al., 2002). SF1 also binds to the spe- observed in Sertoli cells and consequently these transcrip- cific consensus site of the SRY gene, namely nuclear tion factors are thought not to be involved in the AMH hormone receptor 1 (NHR1) to activate the SRY gene dur- expression onset (De Santa Barbara et al., 2000). To spec- ing the gonadal determination pathway (de Santa Barbara ify the targets of WT1 in the gonad, the gene expression et al., 2001). Interaction between SRY and SF1 provides profile was identified in the urogenital ridges of wild type upregulation of Sox9. In addition, the synthesis of cyto- and Wt1 knockout mice using a microarray. Results have chrome p450 enzymes that are necessary in the produc- shown that alteration in the expression level of Wt1 had tion of steroid hormones is known to be regulated by SF1 no effect on Gata4 expression; therefore, Gata4 has been (Wong et al., 1996). reported to be indirectly regulated by Wt1 (Klattig et al., Tissue-specific expression of NR5A1 starts with the for- 2007). mation of the bipotential gonad primordia. Afterward, this process proceeds in the somatic cells of the sex cords and WT1 Leydig cells (Hanley et al., 1999; Achermann et al., 2002). WT1 (OMIM * 607102) is a gene with ten exons encoding SF1 and SOX9 induce the secretion of anti-Mullerian more than 24 different transcription factor isoforms, due 330 MOLECULAR MECHANISMS IN SEX DETERMINATION

TABLE 2. Genes Involved in Sex Determination Process

Association Gene Location MIM number Function(s) in sex determination process with DSDs References

SRY Yp11.3 480000 Promotes male sex determination 1 Sinclair et al. (1990)

Induces SOX9 expression

SOX9 17q24.3 608160 Induces Sertoli cell differentiation 1 Eggers et al. (2014),

Enhances Amh Expression with SF1 Hanley et al. (2000)

NR5A1 9q33 187457 Regulation of steroidogenesis 1 Achermann et al. (2002)

SOX8 16p13.3 605923 Testis cord formation 1 Chaboissier et al. (2004),

Induces the activities of the Otake and Kuroiwa (2016) AMH promoter and TESCO

Consolidates Sox9 function with SF1

SOX10 22q13.1 602229 Consolidates Sox9 function with SF1 1 Chaboissier et al. (2004),

Induces the activities of the Otake and Kuroiwa (2016) AMH promoter and TESCO

SOX3 Xq27.1 313430 Essential for gametogenesis 1 Sutton et al. (2011), and gonadal function Weiss et al. (2003)

Transactivate TESCO with SF1

GATA4 8p23.1 600576 Regulation of Sertoli cell function NA Mazaud-Guittot et al. (2014)

Gonadal functions and development

WT1 11p13 607102 Activation of SRY 1 Hossain and Saunders (2001) FOG2 8q23.1 603693 Regulation of Sertoli cell function 1 Imai et al. (2004), Tevosian et al. (2002)

Gonadal functions and development

NA, not applicable. to alternative splicing and translation start sites and RNA necessary for expression of SOX8 and SOX9 (Gao et al., editing. According to the presence or absence of lysine- 2006). Overexpression or inactivation of Wt1 has resulted threonine-serine (KTS) amino acids between the third and in changes of the AMH type II receptor (Amhr2) expres- fourth zinger fingers, WT1 isoforms are divided into two sion, which plays a role in the regression of the Mullerian groups (KTS1 and KTS2) (Parker et al., 1999; Miyamoto ducts (Klattig et al., 2007). et al., 2008). WT1-KTS isoform and SF1 synergism have been demonstrated to promote AMH expression, and mis- GATA4 sense mutations in WT1 inhibit this synergism and cause GATA4 (OMIM *600576) is a member of GATA-binding pro- to in Denys-Drash syndrome teins recognizing the GATA motif, (A/T)GATA(A/G), and (Nachtigal et al., 1998). WT1 mutations have been known encoded by GATA4 gene that is mapped on 8p23.1. GATA4 to be also involved in Fraiser syndrome, urogenital defect, mRNA is expressed in the heart, stomach, liver, pancreas, and Wilms Tumor-Aniridia-Genitourinary Anomalies-Intel- and yolk sac as well as gonads (Arceci et al., 1993; Xuan and lectual Disability Syndrome (Hossain and Saunders, 2001). Sussel, 2016). GATA4 is involved in various biological func- The Wt1 gene is crucial in multiple steps during testic- tions such as gonadal development and function (Hu et al., ular development. Abnormalities of developing seminifer- 2013; Mazaud-Guittot et al., 2014), regulation of cellular dif- ous tubules and Sertoli cells have been reported in a ferentiation, embryonic morphogenesis (Ketola et al., 2000), mouse strain with Wt1 knock-out allele (Gao et al., 2006). and maintaining pancreas identity (Xuan and Sussel, 2016). SRY activation mediated by WT1 results in the initiation of Friend of GATA2 (FOG2) is a co-factor of GATA4, and a gene cascade during the sex determination process (Hos- both FOG2 and GATA4 take part in critical roles in the sain and Saunders, 2001). Once SRY expression ceases, gonadal development and regulation of Sertoli cell function Wt1 controls Sox9 expression directly or indirectly, and (Tevosian et al., 2002; Imai et al., 2004). Knock-in mutation Wt1 inactivation causes inhibition of AMH, SOX8 and SOX9 in Gata4 (Gata4tm/sho/ Gata4tm/sho) caused failure of the expression in Sertoli cells, suggesting Wt1 is also interaction between GATA4 and FOG2. In normal XY mice, BIRTH DEFECTS RESEARCH (PART C) 108:321–336 (2016) 331

Sry expression is at a maximum level at E11.5 in embryonic was suggested to be connected with demethylation of the development. Yet, in Fog22/2 gonads, SRY transcript levels CpG islands in the Sry locus (Nishino et al., 2011). Hyper- have been found to be decreased due to disturbances in the methylation of the Sry gene causing abnormal gene expres- GATA4/FOG2 interaction (Tevosian et al., 2002; Guittot et al., sion resulted in the silencing of genes, DMRT1, SOX8, SOX9, 2007). The Gata4 promoter includes two conserved box ele- NR5A1, and AMH, involved in the XYDSD gonad in dogs. ments which are G-C and E-box motifs and are necessary for More recently, incomplete demethylation of Sry gene has the activation of Gata4. G-C box motif has a binding site for been suggested to be one of the causes of XYDSD in these SF1 (Guittot et al., 2007). Cooperation of GATA4 and SF1 has dogs (Jeong et al., 2016). been reported to activate the AMH promoter. The SF1 G35E HISTONE MODIFICATIONS mutation has been proposed to cause abnormality in human Nucleosomes are fundamental and functional units of sex determination (Tremblay and Viger, 2003). chromatin, which are composed of histone octamers THE ROLE OF EPIGENETIC MECHANISMS IN SEX DETERMINATION including two copies of each core histone proteins H2A, Epigenetics is currently defined as the modification of phe- H2B, H3 and H4 (Annunziato, 2008). Covalent modifica- notypic characters, without any change in DNA sequence, tions occurring at the N-terminal tails of these histones inherited mitotically and meiotically (Tollefsbol, 2014). are very crucial in gene regulation. There are several kinds The primary epigenetic mechanisms are DNA methylation, of histone tail modifications including methylation, acetyla- histone modifications, and non-coding RNAs (ncRNSs) tion, phosphorylation and ubiquitination (Berger, 2002). (Gibney and Nolan, 2010), mechanisms that are involved Histone H3 Lysine 9 (H3K9) methylation conserved in in the maintenance of gene expression. Epigenetic regula- a variety of species is one of the well-characterized cova- tion of gene expression provides for differentiation of cells, lent modifications. This reversible modification has been obtaining an identity and maintaining its continuity during associated with transcriptional silencing and repressed developmental processes. It is also vital for the sex deter- chromatin state. Furthermore, methylation and demethyla- mination process (Piferrer, 2013). Figure 2 summarizes tion of H3K9 have been proposed to be involved in the the main epigenetic mechanisms in the sex determination regulation of genes associated with sex determination process. (Fuks, 2005, Tachibana, 2015). Jumonji Domain-Containing Protein 1A (JMJD1A) is an enzyme catalyzing demethyla- DNA METHYLATION tion of H3K9. Jmjd1a mutation has caused disturbances in A well-characterized epigenetic mechanism is DNA methyl- Sry gene expression and XY sex reversal (Tachibana, ation that regulates multiple cellular processes such as 2015). genomic imprinting, X chromosome inactivation and Acetylation and deacetylation are the second type of embryonic development (Robertson, 2005). DNA methyla- the histone modifications, which is characterized by trans- tion generally occurs in CpG islands which are character- fer of acetyl groups by two important enzymes called his- ized by over 200 bases with 50% of G1C content. An tone acetyltransferase (HAT) and histone deacetylase association between gene expression and changes in meth- (HDAC). Acetylation removes the positive charge of histone ylation level in the CpG islands has been reported. The tails and leads to loose and relaxed chromatin structure process is catalyzed by enzymes called DNA methyltrans- that allow high levels transcription. Removal of acetyl ferases (DNMTs) which are responsible for the transfer of groups from the lysine (K) residues of histone proteins a methyl group (CH3) from S-adenosyl methionine to 5’ C regulates transcription and chromatin structure; whereas, of cytosine in DNA. DNMT1, DNMT2, DNMT3a, DNMT3b, removal of these acetyl groups from non-histone proteins and DNMT3L have been reported to be included in the regulates various cellular processes (Piferrer, 2013, Seto DNMT family; however, DNMT2 and DNMT3L have no and Yoshida, 2014). On the other hand, acetylation of tran- methyltransferase activity (Piferrer, 2013, Portela and scription factors has been indicated to cause an increase Esteller, 2010). in sequence-specific DNA binding, suggesting that acetyla- DNA methylation plays an indispensable role in cellular tion controls gene function (Sim et al., 2008). differentiation and Sry gene regulation. During embryonic E1A-associated p300 acetyltransferase is one of the development, most (75%) of the 5’ flanking control region nuclear HATs and plays an essential function in the regula- of the Sry gene was methylated at 8.5 d.p.c. Therefore, Sry tion of transcription factor activities through direct inter- expression was not observed. However, in 11.5 d.p.c action and acetylation. During mammalian sex gonads, Sry expression reaches its peak by reducing the determination, SRY activity has been proposed to be regu- methylation state of the 5’ flanking region. Thus, it has lated by acetylation and deacetylation of SRY. Thus, acety- been implicated that Sry expression is inversely associated lation and deacetylation of SRY can give rise to different with the methylation state of Sry (Nishino et al., 2004). outcomes. While SRY acetylation on the lysine 136 residue Methylation of the non-CpG parts of the genome was also (K136) has led to an increase in the interaction between reported and methylation of the CCTGG (non-CpG) region SRY and importin b, which causes nuclear sublocalization 332 MOLECULAR MECHANISMS IN SEX DETERMINATION

of SRY, deacetylation by HDAC3 stimulates cytoplasmic However, the functions of these RNAs in the human sex localization (Thevenet et al., 2004). p300 acetyltransferase determination are still not known. has been reported to activate target genes of Sox9-related transcription factors during the chondrocyte differentiation Conclusions and Perspectives process (Furumatsu and Asahara, 2010). However, there Sex determination is a very complex process and multiple are no data regarding acetylation or deacetylation of SOX9 genes, especially SRY, have crucial functions and are effec- in sex determination and differentiation process. tive in this process. In mammals, a new sex-determining

NON-CODING RNAS gene has recently been reported to activate Sox9 without The ncRNAs are functional RNA molecules that are able to SRY expression (Otake and Kuroiwa, 2016). This indicates manage certain epigenetic alterations including histone that there may be various unknown genes involved in sex modifications and methylation of DNA in complex organ- determination. Yet, recent studies have reported consider- isms. The ncRNAs consist of small non-coding RNAs able roles of epigenetic regulation in the gonadal determi- (sncRNAs) and long non-coding RNAs (lncRNAs) depend- nation (Tachibana, 2015). However, little is known about ing on not only their length, but also their structure and the epigenetic mechanisms and their mode of action. Bio- function. LncRNAs’ length is approximately ten times larg- informatic analyses and advanced molecular techniques er than that of the sncRNAs’, over 200 nucleotides and 20- may aid to clarify the effects of epigenetic mechanisms on 30 nucleotides respectively (Costa, 2008, Liu et al., 2015, sex determination. Therefore, additional studies are need- Piferrer, 2013). LncRNAs, which are newly defined regula- ed to discover new genes or pathways. tory elements, have been reported to be involved in sex determination. In Drosophila lncRNAs were shown to pro- References mote Sxl gene, a master-switch gene in sex determination Achermann JC, Domenice S, Bachega TA, et al. 2015. Disorders (Mulvey et al., 2014). of sex development: effect of molecular diagnostics. Nat Rev Small RNAs including miRNA, siRNA, tRNA, snRNA, Endocrinol 11:478–488. rRNA, and piRNA are functional in various gene regulation Achermann JC, Ito M, Ito, M, et al. 1999. A mutation in the gene processes (Liu et al., 2015). miRNAs regulate gonadal encoding steroidogenic factor-1 causes XY sex reversal and adrenal failure in humans. Nat Genet 22:125–126. functions in gametogenesis, steroidogenesis and germ cell differentiation processes in mammals (Miao et al., 2016, Achermann JC, Ozisik G, Ito M, et al. 2002. Gonadal determina- tion and adrenal development are regulated by the orphan Wang et al., 2016). Differences in miRNA expression pat- nuclear receptor steroidogenic factor-1, in a dose-dependent terns in sex differentiation and dimorphism have been manner. J Clin Endocrinol Metab 87:1829–1833. reported in various species including fish (Xiao et al., Andonova S, Robeva R, Sirakov M, et al. 2015. A novel SRY gene 2014), mouse (Mishima et al., 2008) and chicken. The mutation p.F109L in a 46,XY female with complete gonadal dys- gene expression profiles of miRNAs in chicken gonads at genesis. Sex Dev 9:333–337. E5.5 revealed that twelve miRNAs including miR-107, miR- Annunziato A. 2008. DNA packaging: nucleosomes and chroma- 1b, miR-1694, miR-1570, miR-199, miR-3534 and miR- tin. Nat Educ 1:26. 1767 abounded in male gonads, whereas miR-1607, miR- Arceci RJ, King A, Simon MC, et al. 1993. Mouse GATA-4: a retinoic 181a, miR-7b, miR-218, miR-1637 and miR-715 abounded acid-inducible GATA-binding transcription factor expressed in endo- in female gonads. Moreover, overexpression of miR-107 in dermally derived tissues and heart. Mol Cell Biol 13:2235–2246. male gonads was found to inhibit NR5a1 and p450 family Assumpc¸~ao JG, Benedetti CE, Maciel-Guerra AT, et al. 2002. Novel 19 subfamily A (CYP19A1) gene expression which is the mutations affecting SRY DNA-binding activity: the HMG box downstream target of NR5a1 (Miao et al., 2016). N65H associated with 46,XY pure gonadal dysgenesis and the Liu and colleagues (2015) analyzed small RNA profiles familial non-HMG box R30I associated with variable phenotypes. during gonad reversal from female-specific gonad to male- J Mol Med (Berl) 80:782–790. specific gonad through ovatestis in fish and obtained 47 Baetens D, Stoop H, Peelman F, Mendonc¸a BB, Verdin H, Cools M, million reads of which expressed almost 25% in the ovary, De Baere E. NR5A1 is a novel disease gene for 46,XX testicular 28% in ovatestis and 47% in testis using deep sequencing. and ovotesticular disorders of sex development. Genet Med Clin Genet. 2016 Nov 1. doi:10.1111/cge.12911. [Epub ahead of Most of the piRNAs and miRNAs including 231 conserved print] Review. PMID:27801941. and 1000 novel miRNAs differentially expressed during Barbaro M, Cools M, Looijenga L, et al. 2011. Partial deletion of the gonad reversal (Liu et al., 2015). In mouse, expression lev- NR5A1 (SF1) gene detected by synthetic probe MLPA in a patient els of miR-202-5p and miR-202-3p were regulated by Sox9 with XY gonadal disorder of sex development. Sex Dev 5:181–187. during sex determination and differentiation processes Barrionuevo F, Georg I, Scherthan H, et al. 2009. Testis cord dif- (Wainwright et al., 2013). Studies conducted in various ferentiation after the sex determination stage is independent of animal models have revealed the importance of ncRNAs Sox9 but fails in the combined absence of Sox9 and Sox8. Dev during gonadal determination and differentiation process. Biol 327:301–312. BIRTH DEFECTS RESEARCH (PART C) 108:321–336 (2016) 333

Barrionuevo F, Scherer G. 2010. SOX E genes: SOX9 and SOX8 in Costa FF. 2008. Non-coding RNAs, epigenetics and complexity. mammalian testis development. Int J Biochem Cell Biol 42:433–436. Gene 410:9–17. Barrionuevo FJ, Burgos M, Scherer G, Jimenez R. 2012. Genes Cotinot C, Pailhoux E, Jaubert F, Fellous M. 2002. Molecular promoting and disturbing testis development. Histol Histopathol genetics of sex determination. Seminars in reproductive medi- 27:1361–1383. cine. New York, NY: Thieme Medical Publishers. Barrionuevo FJ, Hurtado A, Kim GJ, et al. 2016. Sox9 and Sox8 De Santa Barbara P, Mejean C, Moniot B, et al. 2001. Steroidogen- protect the adult testis from male-to-female genetic reprogram- ic factor-1 contributes to the cyclic-adenosine monophosphate ming and complete degeneration. Elife 5. PMID:27328324 PMI- down-regulation of human SRY gene expression. Biol Reprod 64: CID: PMC4945155, DOI:10.7554/eLife.15635. 775–783. Bashamboo A, Donohoue PA, Vilain E, Eozenou C, Rojo S, De Santa Barbara P, Moniot B, Poulat F, Berta, P. 2000. Expres- McElreavey K. A recurrent p.Arg92Trp variant in steroidogenic sion and subcellular localization of SF-1, SOX9, WT1, and AMH factor-1 (NR5A1) can act as a molecular switch in human sex devel- proteins during early human testicular development. Dev Dyn opment. Hum Mol Genet Clin Genet. 2016 Nov 28. doi:10.1111/ 217:293–298. cge.12932. [Epub ahead of print] Review. PMID:27893151. Domenice S, Yumie Nishi M, Correia Billerbeck AE, et al. 1998. A Battiloro E, Angeletti B, Tozzi MC, et al. 1997. A novel double novel missense mutation (S18N) in the 5’ non-HMG box region nucleotide substitution in the HMG box of the SRY gene associat- of the SRY gene in a patient with partial gonadal dysgenesis and ed with Swyer syndrome. Hum Genet 100:585–587. his normal male relatives. Hum Genet 102:213–215. Benko S, Gordon CT, Mallet D, et al. 2011. Disruption of a long Eggers S, Ohnesorg T, Sinclair, A. 2014. Genetic regulation distance regulatory region upstream of SOX9 in isolated disor- of mammalian gonad development. Nat Rev Endocrinol 10:673– ders of sex development. J Med Genet 48:825–830. 683. Berger SL. 2002. Histone modifications in transcriptional regula- Fabbri HC, De Andrade JGR, Soardi FC, et al. 2014. The novel p. tion. Curr Opin Genet Dev 12:142–148. Cys65Tyr mutation in NR5A1 gene in three 46, XY siblings with Berta P, Hawkins JB, Sinclair AH, et al. 1990. Genetic evidence equat- normal levels and their mother with primary ovari- ing SRY and the testis-determining factor. Nature 348:448–450. an insufficiency. BMC Med Genet 15:1. Bi W, Deng JM, Zhang Z, et al. 1999. Sox9 is required for carti- Filges I, Kunz C, Miny P, et al. 2011. A novel missense mutation lage formation. Nat Genet 22:85–89. in the high mobility group domain of SRY drastically reduces its DNA-binding capacity and causes paternally transmitted 46,XY Bilbao JR, Loridan L, Castano~ L. 1996. A novel postzygotic non- sense mutation in SRY in familial XY gonadal dysgenesis. Hum complete gonadal dysgenesis. Fertil Steril 96:851–855. Genet 97:537–539. Foster JW, Dominguez-Steglich MA, Guioli S, et al. 1994. Campo- Britsch S, Goerich DE, Riethmacher D, et al. 2001. The transcrip- melic dysplasia and autosomal sex reversal caused by mutations tion factor Sox10 is a key regulator of peripheral glial develop- in an SRY-related gene. Nature 372:525–530. ment. Genes Dev 15:66–78. Foster JW, Graves J. 1994. An SRY-related sequence on the Bylund M, Andersson E, Novitch BG, Muhr J. 2003. Vertebrate marsupial X chromosome: implications for the evolution of the neurogenesis is counteracted by Sox1–3 activity. Nat Neurosci 6: mammalian testis-determining gene. Proc Natl Acad Sci USA 91: 1162–1168. 1927–1931. Canto P, de la Chesnaye E, Lopez M, et al. 2000. A mutation in Fuks F. 2005. DNA methylation and histone modifications: the 5’ non-high mobility group box region of the SRY gene in teaming up to silence genes. Curr Opin Genet Dev 15:490–495. patients with Turner syndrome and Y mosaicism. J Clin Endocri- Furumatsu T, Asahara H. 2010. Histone acetylation influences the nol Metab 85:1908–1911. activity of Sox9-related transcriptional complex. Acta Med Okaya- Chaboissier MC, Kobayashi A, Vidal VI, et al. 2004. Functional ma 64:351–357. analysis of Sox8 and Sox9 during sex determination in the Gao F, Maiti S, Alam N, et al. 2006. The Wilms tumor gene, Wt1, mouse. Development 131:1891–1901. is required for Sox9 expression and maintenance of tubular Chaoui A, Kavo A, Baral V, et al. 2015. Subnuclear re-localization architecture in the developing testis. Proc Natl Acad Sci USA of SOX10 and p54NRB correlates with a unique neurological 103:11987–11992. phenotype associated with SOX10 missense mutations. Hum Mol Georg I, Barrionuevo F, Wiech T, Scherer G. 2012. Sox9 and Sox8 Genet 24:4933–4947. are required for basal lamina integrity of testis cords and for Cheung M, Briscoe J. 2003. Neural crest development is regulated suppression of FOXL2 during embryonic testis development in by the transcription factor Sox9. Development 130:5681–5693. mice. Biol Reprod 87:99. Clepet C, Schater AJ, Sinclair AH, et al. 1993. The human SRY Gerhardt K. 2016. World of reproductive biology: the fate of sex: transcript. Hum Mol Genet 2:2007–2012. SRY on the ridge. Biol Reprod. The Fate of Sex: SRY on the Ridge 95:106. PMID:27856452. Collignon J, Sockanathan S, Hacker A, et al. 1996. A comparison of the properties of Sox-3 with Sry and two related genes, Sox-1 Gibney E, Nolan C. 2010. Epigenetics and gene expression. and Sox-2. Development 122:509–520. Heredity 105:4–13. Cory AT, Boyer A, Pilon N, et al. 2007. Presumptive pre-Sertoli Gordon CT, Attanasio C, Bhatia S, et al. 2014. Identification of cells express genes involved in cell proliferation and cell signal- novel craniofacial regulatory domains located far upstream of ing during a critical window in early testis differentiation. Mol SOX9 and disrupted in Pierre Robin sequence. Hum Mutat 35: Reprod Dev 74:1491–1504. 1011–1020. 334 MOLECULAR MECHANISMS IN SEX DETERMINATION

Guittot SM, Tetu A, Legault E, et al. 2007. The proximal Gata4 Jeong Y-H, Lu H, Park C-H, et al. 2016. Stochastic anomaly of promoter directs reporter gene expression to sertoli cells during methylome but persistent SRY hypermethylation in disorder of mouse gonadal development. Biol Reprod 76:85–95. sex development in canine somatic cell nuclear transfer. Sci Rep 6. Gunes S, Asci R, Okten G, et al. 2013. Two males with SRY- Jordan BK, Jain M, Natarajan S, et al. 2002. Familial mutation in positive 46,XX testicular disorder of sex development. Syst Biol the testis-determining gene SRY shared by an XY female and her Reprod Med 59:42–47. normal father. J Clin Endocrinol Metab 87:3428–3432. Haines B, Hughes J, Corbett M, et al. 2015. Interchromosomal Katoh-Fukui Y, Igarashi M, Nagasaki K, et al. 2015. Testicular dys- insertional translocation at Xq26.3 alters SOX3 expression in an genesis/regression without in patients car- individual with XX male sex reversal. J Clin Endocrinol Metab rying missense mutations and upstream deletion of SOX9. Mol 100, E815–20. Genet Genomic Med 3:550–357. Hanley N, Ball S, Clement-Jones M, et al. 1999. Expression of ste- Kent J, Wheatley SC, Andrews JE, et al. 1996. A male-specific role roidogenic factor 1 and Wilms’ tumour 1 during early human for SOX9 in vertebrate sex determination. Development 122: gonadal development and sex determination. Mech Dev 87:175– 2813–2822. 180. Ketola I, Pentika€inen V, Vaskivuo T, et al. 2000. Expression of Hanley N, Hagan D, Clement-Jones M, et al. 2000. SRY, SOX9, and transcription factor GATA-4 during human testicular develop- DAX1 expression patterns during human sex determination and ment and disease 1. J Clin Endocrinol Metab 85:3925–3931. gonadal development. Mech Dev 91:403–407. Kim, GJ, Sock E, Buchberger A, et al. 2015. Copy number varia- Harley VR, Layfield S, Mitchell CL, et al. 2003. Defective importin tion of two separate regulatory regions upstream of SOX9 causes beta recognition and nuclear import of the sex-determining fac- isolated 46,XY or 46,XX disorder of sex development. J Med tor SRY are associated with XY sex-reversing mutations. Proc Genet 52:240–247. Natl Acad Sci USA 100:7045–7050. Kimura R, Murata C, Kuroki Y, Kuroiwa A. 2014. Mutations in the Hawkins JR, Taylor A, Berta P, et al. 1992a. Mutational analysis testis-specific enhancer of SOX9 in the SRY independent sex- of SRY: nonsense and missense mutations in XY sex reversal. determining mechanism in the genus Tokudaia. PLoS One 9: Hum Genet 88:471–474. e108779. Hawkins JR, Taylor A, Goodfellow PN, et al. 1992b. Evidence for Klattig J, Sierig R, Kruspe D, et al. 2007. WT1-mediated gene reg- increased prevalence of SRY mutations in XY with com- ulation in early urogenital ridge development. Sex Dev 1:238– plete rather than partial gonadal dysgenesis. Am J Hum Genet 254. 51:979–984. Kojima Y, Hayashi Y, Mizuno K, et al. 2008. Up-regulation of Helszer Z, Dmochowska A, Szemraj J, et al. 2013. A novel muta- SOX9 in human sex-determining region on the tion (c. 341A>G) in the SRY gene in a 46,XY female patient with (SRY)-negative XX males. Clin Endocrinol (Oxf) 68:791–799. gonadal dysgenesis. Gene 526:467–470. Kuhlbrodt K, Schmidt C, Sock E, et al. 1998. Functional analysis Herpin A, Schartl M. 2011. Sex determination: switch and sup- of Sox10 mutations found in human Waardenburg-Hirschsprung press. Curr Biol 21:R656–659. patients. J Biol Chem 273:23033–23038. Hersmus R, de Leeuw BH, Stoop H, et al. 2009. A novel SRY mis- Kuroki S, Matoba S, Akiyoshi M, et al. 2013. Epigenetic regulation sense mutation affecting nuclear import in a 46,XY female patient of mouse sex determination by the histone demethylase Jmjd1a. with bilateral gonadoblastoma. Eur J Hum Genet 17:1642–1649. Science 341:1106–1109. Hossain A, Saunders GF. 2001. The human sex-determining gene Lee YH, Saint-Jeannet JP. 2011. Sox9 function in craniofacial SRY is a direct target of WT1. J Biol Chem 276:16817–16823. development and disease. Genesis 49:200–208. Hu Y-C, Okumura LM, Page DC. 2013. Gata4 is required for for- Liu J, Luo M, Sheng Y, et al. 2015. Dynamic evolution and biogen- mation of the genital ridge in mice. PLoS Genet 9:e1003629. esis of small RNAs during sex reversal. Sci Rep 5:9999. Huang YH, Jankowski A, Cheah KS, et al. 2015. SOXE transcrip- Lucas-Herald A, Bashamboo A. 2014. Hiort O, Ahmed SF (eds): tion factors form selective dimers on non-compact DNA motifs Gonadal development. Understanding Differences and Disorders through multifaceted interactions between dimerization and of Sex Development (DSD). Endocr Dev. Basel, Karger Publishers. high-mobility group domains. Sci Rep 5:10398. 27:1–16. doi:10.1159/000363608, see https://www.karger.com/ Hughes IA, Houk C, Ahmed SF, et al. 2006. Consensus state- Article/Abstract/363608. PMID:25247640. ment on management of disorders. J Pediatr Urol 2: Lybaek H, De Bruijn D, Den Engelsman-Van Dijk AH, et al. 2014. 148–162. RevSex duplication-induced and sex-related differences in the Hyon C, Chantot-Bastaraud S, Harbuz R, et al. 2015. Refining the SOX9 regulatory region chromatin landscape in human fibro- regulatory region upstream of SOX9 associated with 46,XX testic- blasts. Epigenetics 9:416–27. ular disorders of Sex Development (DSD). Am J Med Genet A Maier EM, Leitner C, Lohrs€ U, Kuhnle U. 2003. True hermaphro- 167A:1851–1858. ditism in an XY individual due to a familial point mutation of the Imai T, Kawai Y, Tadokoro Y, et al. 2004. In vivo and in vitro con- SRY gene. J Pediatr Endocrinol Metab 16:575–580. stant expression of GATA-4 in mouse postnatal Sertoli cells. Mol Matzuk MM, Lamb DJ. 2008. The biology of infertility: research Cell Endocrinol 214:107–115. advances and clinical challenges. Nat Med 14:1197–213. J€ager RJ, Harley VR, Pfeiffer RA, et al. 1992. A familial mutation Mazaud-Guittot S, Prud’homme B, Bouchard MF, et al. 2014. in the testis-determining gene SRY shared by both sexes. Hum GATA4 autoregulates its own expression in mouse gonadal cells Genet 90:350–355. via its distal 1b promoter. Biol Reprod 90:25. BIRTH DEFECTS RESEARCH (PART C) 108:321–336 (2016) 335

McElreavey KD, Vilain E, Boucekkine C, et al. 1992. XY sex rever- Pfeifer D, Poulat F, Holinski-Feder E, et al. 2000. The SOX8 gene is sal associated with a nonsense mutation in SRY. Genomics 13: located within 700 kb of the tip of chromosome 16p and is delet- 838–840. ed in a patient with ATR-16 syndrome. Genomics 63:108–116. Miao N, Wang X, Hou Y, et al. 2016. Identification of male-biased Philibert P, Leprieur E, Zenaty D, et al. 2010. Steroidogenic microRNA-107 as a direct regulator for nuclear receptor subfam- factor-1 (SF-1) gene mutation as a frequent cause of primary ily 5 group A member 1 based on sexually dimorphic microRNA amenorrhea in 46, XY female adolescents with low testosterone expression profiling from chicken embryonic gonads. Mol Cell concentration. Reprod Biol Endocrinol 8:1. Endocrinol 429:29–40. Piferrer F. 2013. Epigenetics of sex determination and gonado- Mishima T, Takizawa T, Luo SS, et al. 2008. MicroRNA (miRNA) genesis. Dev Dyn 242:360–370. cloning analysis reveals sex differences in miRNA expression pro- Pingault V, Bondurand N, Kuhlbrodt K, et al. 1998. SOX10 muta- files between adult mouse testis and ovary. Reproduction 136: tions in patients with Waardenburg-Hirschsprung disease. Nat 811–22. Genet 18:171–173. Mitchell CL, Harley VR. 2002. Biochemical defects in eight SRY Polanco JC, Wilhelm D, Davidson TL, et al. 2010. Sox10 gain-of- missense mutations causing XY gonadal dysgenesis. Mol Genet function causes XX sex reversal in mice: implications for human Metab 77:217–225. 22q-linked disorders of sex development. Hum Mol Genet 19: Miyamoto Y, Taniguchi H, Hamel F, et al. 2008. A GATA4/WT1 coop- 506–516. eration regulates transcription of genes required for mammalian Pompolo S, Harley V. 2001. Localisation of the SRY-related HMG sex determination and differentiation. BMC Mol Biol 9:1. box protein, SOX9, in rodent brain. Brain Res 906:143–148. Moalem S, Babul-Hirji R, Stavropolous DJ, et al. 2012. XX male Portela A, Esteller M. 2010. Epigenetic modifications and human sex reversal with genital abnormalities associated with a de disease. Nat Biotechnol 28:1057–1068. novo SOX3 gene duplication. Am J Med Genet A 158A:1759– 1764. Poulat F, Desclozeaux M, Tuffery S, et al. 1998. Mutation in the 5’ noncoding region of the SRY gene in an XY sex-reversed Mulvey BB, Olcese U, Cabrera JR, Horabin JI. 2014. An interactive patient. Hum Mutat (Suppl 1):S192–S194. network of long non-coding RNAs facilitates the Drosophila sex determination decision. Biochim Biophys Acta 1839:773–784. Premi S, Srivastava J, Chandy SP, et al. 2006. Tandem duplication and copy number polymorphism of the SRY gene in patients Nachtigal MW, Hirokawa Y, Enyeart-Vanhouten DL, et al. 1998. with sex chromosome anomalies and males exposed to natural Wilms’ tumor 1 and Dax-1 modulate the orphan nuclear receptor background radiation. Mol Hum Reprod 12:113–121. SF-1 in sex-specific gene expression. Cell 93:445–454. Pusch C, Hustert E, Pfeifer D, et al. 1998. The SOX10/Sox10 gene Nishino K, Hattori N, Sato S, et al. 2011. Non-CpG methylation from human and mouse: sequence, expression, and transactiva- occurs in the regulatory region of the Sry gene. J Reprod Dev 57: tion by the encoded HMG domain transcription factor. Hum 586–593. Genet 103:115–123. Nishino K, Hattori N, Tanaka S, Shiota K. 2004. DNA methylation- Racca JD, Chen Y-S, Yang Y, et al. 2016. Human sex determination mediated control of Sry gene expression in mouse gonadal devel- at the edge of ambiguity inherited XY sex reversal due to opment. J Biol Chem 279:22306–22313. enhanced ubiquitination and proteasomal degradation of a mas- Oba K, Yanase T, Nomura M, et al. 1996. Structural Characteriza- ter transcription factor. J Biol Chem 291:22173–22195. tion of humanAd4bp (SF-1) Gene. Biochem Biophys Res Commun Rigby N, Kulathinal RJ. 2015. Genetic architecture of sexual 226:261–267. dimorphism in humans. J Cell Physiol 230:2304–2310. O’Bryan MK, Takada S, Kennedy CL, et al. 2008. Sox8 is a critical Rizzoti K, Brunelli S, Carmignac D, et al. 2004. SOX3 is required during regulator of adult Sertoli cell function and male fertility. Dev Biol the formation of the hypothalamo-pituitary axis. Nat Genet 36:247–255. 316:359–370. Robertson KD. 2005. DNA methylation and human disease. Nat Ohnesorg T, Vilain E, Sinclair AH. 2014. The genetics of disorders Rev Genet 6:597–610. of sex development in humans. Sex Dev 8:262–272. Ronfani L, Bianchi M. 2004. Molecular mechanisms in male Ortega EA, Ruthig VA, Ward MA. 2015. Sry-independent overex- determination and germ cell differentiation. Cellular and Molecu- pression of Sox9 supports spermatogenesis and fertility in the lar Life Sciences CMLS, 61:1907–1925. mouse. Biol Reprod. 93:141. Sadler TW. 2011. Langman’s medical embryology. Thomas W. Otake T, Kuroiwa A. 2016. Molecular mechanism of male Sadler 12th edition Philadelphia, PA. Lippincott Williams & differentiation is conserved in the SRY-absent mammal, Tokudaia Wilkins. osimensis. Sci Rep 6. Sch€affler A, Barth N, Winkler K, et al. 2000. Identification of a Page DC, Mosher R, Simpson EM, et al. 1987. The sex-determining new missense mutation (Gly95Glu) in a highly conserved region of the human Y chromosome encodes a finger protein. Cell codon within the high-mobility group box of the sex-determin- 51:1091–1104. ing region Y gene: report on a 46,XY female with gonadal dys- Paliwal P, Sharma A, Birla S, et al. 2011. Identification of novel genesis and yolk-sac tumor. J Clin Endocrinol Metab 85:2287– SRY mutations and SF1 (NR5A1) changes in patients with pure 2292. gonadal dysgenesis and 46,XY karyotype. Mol Hum Reprod 17: Schepers G, Wilson M, Wilhelm D, Koopman P. 2003. SOX8 is 372–378. expressed during testis differentiation in mice and synergizes Parker KL, Schimmer BP, Schedl A. 1999. Genes essential for ear- with SF1 to activate the Amh promoter in vitro. J Biol Chem ly events in gonadal development. Cell Mol Life Sci 55:831–838. 278:28101–28108. 336 MOLECULAR MECHANISMS IN SEX DETERMINATION

Scherer G, Held M, Erdel M, et al. 1998. Three novel SRY muta- Egyptian patients with 46, XY disorders of sex development. Eur tions in XY gonadal dysgenesis and the enigma of XY gonadal J Endocrinol 170:759–767. dysgenesis cases without SRY mutations. Cytogenet Cell Genet Tevosian SG, Albrecht KH, Crispino JD, et al. 2002. Gonadal dif- 80:188–192. ferentiation, sex determination and normal Sry expression in Schlessinger D, Garcia-Ortiz JE, Forabosco A, et al. 2010. Determi- mice require direct interaction between transcription partners nation and stability of gonadal sex. J Androl 31:16–25. GATA4 and FOG2. Development 129:4627–4634. Sekido R, Lovell-Badge R. 2008. Sex determination involves syn- Thevenet L, Mejean C, Moniot B, et al. 2004. Regulation of ergistic action of SRY and SF1 on a specific Sox9 enhancer. human SRY subcellular distribution by its acetylation/deacetyla- Nature 453:930–4. tion. EMBO J 23:3336–3345. Seto E, Yoshida M. 2014. Erasers of histone acetylation: the his- Tollefsbol T. 2014. Transgenerational epigenetics. Trygve Tollefs- tone deacetylase enzymes. Cold Spring Harbor Persp Biol 6: bol, 1st edition, San Diego, Elsevier. a018713. Tremblay JJ, Viger RS. 2003. A mutated form of steroidogenic factor Shahid M, Dhillon VS, Aslam M, Husain SA. 2005. Three new 1 (SF-1 G35E) that causes sex reversal in humans fails to synergize novel point mutations localized within and downstream of with transcription factor GATA-4. J Biol Chem 278:42637–42642. high-mobility group-box region in SRY gene in three Indian Veitia R, Ion A, Barbaux S, et al. 1997. Mutations and sequence females with Turner syndrome. J Clin Endocrinol Metab 90: variants in the testis-determining region of the Y chromosome in 2429–2435. individuals with a 46,XY female phenotype. Hum Genet 99:648– Shahid M, Dhillion VS, Jain N, et al. 2004. Two new novel point 652. mutations localized upstream and downstream of the HMG box Vidal VP, Chaboissier M-C, De Rooij DG, Schedl A. 2001. Sox9 region of the SRY gene in three Indian 46,XY females with sex induces testis development in XX transgenic mice. Nat Genet 28: reversal and gonadal tumour formation. Mol Hum Reprod 10: 216–217. 521–526. Wagner T, Wirth J, Meyer J, et al. 1994. Autosomal sex reversal She ZY, Yang WX. Sry and SoxE genes: how they participate in and campomelic dysplasia are caused by mutations in and mammalian sex determination and gonadal development? Semin around the SRY-related gene SOX9. Cell 79:1111–1120. Cell Dev Biol 2016 Jul 29. pii: S1084-9521(16)30236-1. doi:10.1016/j.semcdb.2016.07.032. [Epub ahead of print] Wainwright EN, Jorgensen JS, Kim Y, et al. 2013. SOX9 regulates microRNA miR-202-5p/3p expression during mouse testis differ- Shen W-H, Moore CC, Ikeda Y, et al. 1994. Nuclear receptor ste- entiation. Biol Reprod 89:34. roidogenic factor 1 regulates the Mullerian€ inhibiting substance gene: a link to the sex determination cascade. Cell 77:651–661. Wang W, Liu W, Liu Q, et al. 2016. Coordinated microRNA and messenger RNA expression profiles for understanding sexual Sim H, Argentaro A, Harley VR. 2008. Boys, girls and shuttling of dimorphism of gonads and the potential roles of microRNA in SRY and SOX9. Trends Endocrinol Metab 19:213–222. the steroidogenesis pathway in Nile tilapia (Oreochromis niloti- Skinner MK, Bhandari RK, Haque MM, Nilsson EE. 2015. Environ- cus). Theriogenology 85:970–978. mentally induced epigenetic transgenerational inheritance of Weiss J, Meeks JJ, Hurley L, et al. 2003. Sox3 is required for altered SRY genomic binding during gonadal sex determination. gonadal function, but not sex determination, in males and Environ Epigenet 1:dvv004. females. Mol Cell Biol 23:8084–8091. Spokony RF, Aoki Y, Saint-Germain N, Magner-Fink E, Saint- Wijchers PJ, Festenstein RJ. 2011. Epigenetic regulation of auto- Jeannet J-P. 2002. The transcription factor Sox9 is required for somal gene expression by sex chromosomes. Trends Genet 27: cranial neural crest development in Xenopus. Development 129: 132–140. 421–432. Wilhelm D, Yang JX, Thomas P. 2013. Mammalian sex determina- Stevanovlc´ M, Lovell-Badge R, Goodfellow PN. 1993. SOX3 is an tion and gonad development. Curr Top Dev Biol 106:89–121. X-linked gene related to SRY. Hum Mol Genet 2:2013–2018. Wong M, Ramayya M, Chrousos G, et al. 1996. Cloning and Stoppa-Vaucher S, Ayabe T, Paquette J, et al. 2012. 46, XY gonadal sequence analysis of the human gene encoding steroidogenic fac- dysgenesis: new SRY point mutation in two siblings with pater- tor 1. J Mol Endocrinol 17:139–147. nal germ line mosaicism. Clin Genet 82:505–513. Xia XY, Zhang C, Li TF, et al. 2015. A duplication upstream of Sutton E, Hughes J, White S, et al. 2011. Identification of SOX3 as SOX9 was not positively correlated with the SRYnegative 46,XX an XX male sex reversal gene in mice and humans. J Clin Invest testicular disorder of sex development: A case report and litera- 121:328–341. ture review. Mol Med Rep 12:5659–5664. Tachibana M. 2015. Epigenetics of sex determination in mam- Xiao J, Zhong H, Zhou Y, et al. 2014. Identification and characteri- mals. Reprod Med Biol 15:59–67. zation of microRNAs in ovary and testis of Nile tilapia (Oreochro- Takagi A, Imai A, Tamaya T. 1999. A novel sex-determining mis niloticus) by using solexa sequencing technology. PLoS One region on Y (SRY) nonsense mutation identified in a 45,X/47,XYY 9:e86821. female. Fertil Steril 72:167–169. Xuan S, Sussel L. 2016. GATA4 and GATA6 regulate pancreatic Taketo M, Parker KL, Howard TA, et al. 1995. Homologs of Dro- endoderm identity through inhibition of hedgehog signaling. sophila Fushi-Tarazu factor 1 map to mouse chromosome 2 and Development 143:780–786. human chromosome 9q33. Genomics 25:565–567. Zangen D, Kaufman Y, Banne E, et al. 2014. Testicular differentia- Tantawy S, Mazen I, Soliman H, et al. 2014. Analysis of the gene tion factor SF-1 is required for human spleen development. coding for (SF1, NR5A1) in a cohort of 50 J Clin Invest 124:2071–2075.