Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Review Article

Sex Dev 2016;10:313–325 Accepted: September 30, 2016


by M. Schmid
DOI: 10.1159/000452637
Published online: December 3, 2016

Mechanism of Sex Determination in


Humans: Insights from Disorders of Sex
Development
Anu Bashamboo Ken McElreavey
Human Developmental Genetics, Department of Development and Stem Cell Biology, CNRS, UMR 3738, Institut
Pasteur, Paris, France

Key Words Disorders of sex development (DSD) are defined as


Ambiguous genitalia · Disorder of sex development · congenital conditions with discordant development of
Gonadal dysgenesis · Mutation chromosomal and gonadal/anatomical sex. Just over 10
years ago, at the Chicago Consensus conference, the term
DSD was coined to include previous descriptions such
Abstract as intersex, pseudohermaphroditism, hermaphroditism,
In this review we will consider the gene mutations respon- and sex reversal [Lee et al., 2006]. These terms were often
sible for the non-syndromic forms of disorders of sex devel- confusing, both to clinicians and patients as well as to
opment (DSD) and how recent genetic findings are provid- other family members. This umbrella definition of DSD
ing insights into the mechanism of sex determination. High- provides a rational basis for the classification of a range
throughput sequencing technologies are having a major of conditions, but more importantly, it avoids confusion
impact on our understanding of the genetic basis of rare hu- with terms such as transgender, gender dysphoria, or ho-
man disorders, including DSD. The study of human DSD is mosexuality.
progressively revealing subtle differences in the genetics of 46,XY DSD includes aberrant testis determination or
the sex-determining system between the mouse and the hu- undermasculinization of an XY male due to errors in ei-
man. This plasticity of the sex-determining pathway is appar- ther androgen synthesis or action. Errors in testis deter-
ent in (a) the difference in phenotypes in human and mouse mination may manifest as either complete gonadal dys-
associated with the same gene, (b) the different gene regula- genesis (CGD) or partial gonadal dysgenesis (PGD).
tory mechanisms between human and mouse, and finally (c) 46,XY CGD is characterized by completely female exter-
the different and unexpected reproductive phenotypes nal genitalia, well-developed Müllerian structures, and a
seen in association with mutations in well-studied sex-deter- gonad composed of a streak of fibrous tissue, whereas
mining genes. © 2016 S. Karger AG, Basel 46,XY PGD is characterized by partial testis formation,
usually a mixture of Wolffian and Müllerian ducts, and
varying degrees of masculinization of the external genita-
lia. Embryonic testicular regression sequence can also be

© 2016 S. Karger AG, Basel Anu Bashamboo


1661–5425/16/0106–0313$39.50/0 Human Developmental Genetics, Institut Pasteur
25 Rue du Dr Roux
E-Mail karger@karger.com
FR–75724 Paris cedex 15 (France)
www.karger.com/sxd
E-Mail anu.bashamboo @ pasteur.fr
regarded as part of the clinical spectrum of 46,XY gonad- viduals that have both ovarian and testicular tissue in the
al dysgenesis [Marcantonio et al., 1994]. Affected indi- gonads, usually ovotestes but less commonly a testis (or
viduals have a 46,XY karyotype and usually present with ovotestis) on one side and an ovary on the other [Ergun-
ambiguous external or internal genitalia. Gonad tissue is Longmire et al., 2005]. The external genitalia are usually
absent on one or both sides. Patients with this condition ambiguous or feminine, with the degree of masculiniza-
are considered to have incomplete testicular determina- tion broadly correlating with the amount of testicular tis-
tion with the loss of gonad material early in gestation be- sue present. In both TDSD and OTDSD the histological
fore testis differentiation is complete [Marcantonio et al., examination of the gonads shows distinct tubule struc-
1994]. This also raises the concept that we are dealing tures in the testicular-like tissue and the presence of fol-
with a continuum of phenotypes rather than clearly dis- licles in the ovarian-like tissue, in the case of OTDSD.
tinct and unrelated categories of atypical testicular for- Sex chromosome DSD includes 47,XXY (Klinefelter
mation. The genetic arguments also favor this, since in syndrome and variants), 45,X (Turner syndrome and
some families with 46,XY DSD, the affected individuals variants), 45,X/46,XY (mixed gonadal dysgenesis or
can present as girls with either CGD or PCG or as men OTDSD), and 46,XX/46,XY (chimerism or OTDSD).
with isolated hypospadias and/or cryptorchidism [Le 45,X/46,XY mosaicism is one of the most common causes
Caignec et al., 2003]. Other forms of 46,XY DSD are dis- of DSD and also one of the most difficult for predicting
orders in androgen synthesis or action. This includes an- genotype-phenotype correlations. In several studies, no
drogen biosynthesis defects such as 17-hydroxysteroid correlation was found between the proportion of the
dehydrogenase deficiency, 5α reductase deficiency, and 45,X/46,XY cell lines in the blood or the fibroblasts of the
StAR mutations. Defects in androgen action include com- patient and the phenotype. Furthermore, some of these
plete or partial androgen insensitivity associated with cases show mild intellectual disability or signs of autism.
mutations in the androgen receptor. Recessive LH recep- A large number of syndrome associations of DSD have
tor mutations result in Leydig cell hypoplasia or aplasia. been described including cloacal anomalies, Robinow,
Central hypogonadotropic hypogonadism (CHH) occurs Aarskog, Hand-Foot-Genital, popliteal pterygium and
when the physiologic function of the hypothalamic-pitu- Serkel syndrome, and Müllerian duct aplasia. This review
itary-gonadal axis is compromised. In 46,XY individuals will focus on recent developments in our understanding
CHH is characterized by delayed or absent sexual devel- of the non-syndromic forms of DSD associated with er-
opment and infertility associated with inappropriately rors in sex determination.
low gonadotropin (LH and FSH) and testosterone levels.
Male patients frequently show under-androgenisation
with micropenis and cryptorchidism observed at birth. Incidence of Disorders of Sex Development
When anomalies of smell, hyposmia or anosmia, is asso-
ciated with hypogonadotropic hypogonadism, in 60% of There is very limited data available on the precise in-
patients the disease is called Kallmann syndrome. This cidence of DSD. This reflects both the rarity of some of
combination of phenotypes is explained by the common these conditions as well as the challenge of achieving a
embryonic origins and developmental pathways of GnRH definitive clinical diagnosis. In the newborn, truly am-
and olfactory neurons. More than 20 genes are known to biguous genitalia that may pose a problem for binary gen-
cause CHH [Marino et al., 2014]. der assignment has an estimated incidence of 1: 4,500–
46,XX DSD includes overvirilization or masculiniza- 5,500 births [Thyen et al., 2006; Sax, 2002]. Overall,
tion of an XX individual due to androgen excess, and the around 50% of all cases of DSD with truly ambiguous gen-
vast majority of cases of 46,XX DSD are due to congen- italia are due to either CAH or 46,XY mixed gonadal dys-
ital adrenal hyperplasia (CAH). The most common form genesis caused by a 45,X/46,XY mosaicism [Thyen et al.,
of CAH is due to deficiency of 21-hydroxylase, which 2006]. The incidence of 46,XY DSD is estimated to be
is caused by mutations in the 21-hydroxylase gene 1:20,000 births and of 46,XY gonadal dysgenesis around
(CYP21A2) and accounts for 90–95% of all cases [White 1:100,000 births [Lee et al., 2016]. TDSD/OTDSD are es-
et al., 1984; Arlt and Krone, 2007]. The much rarer forms timated to occur in 1: 100,000 births [Sax, 2002]. More
are 46,XX testicular DSD (TDSD) and 46,XX ovotesticu- commonly, developmental anomalies of the external gen-
lar DSD (OTDSD). Individuals with TDSD are males italia may exist in 1 in 300 newborn infants [Nordenvall
with small and azoospermic testis [de la Chapelle, 1972] et al., 2014]. These include undescended testis or anoma-
and a normal male habitus. 46,XX OTDSD refers to indi- lies of the opening of the urethra on the penis (hypospa-

314 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
dias). However, most of this published data on the inci- incomplete penetrance could be caused by stochastic ef-
dence of DSD is available only from Western countries; fects around a threshold level of biological activity re-
therefore, the worldwide prevalence of DSD is unclear. A quired for testis formation [Phillips et al., 2011]. Although
German study indicated that the incidence of ambiguous SRY mutations are usually associated with gonadal dys-
genitalia in infants of non-German background was 4× genesis, a 46,XY woman with premature menopause was
higher compared to the general population [Thyen et al., reported to carry a de novo p.Gln2Ter mutation [Brown
2006], which they attributed to an increase in autosomal et al., 1998].
recessive forms of DSD due to higher rates of consanguin- The direct target of SRY is SOX9, another HMG-box
ity in the migrant populations. There is some evidence to containing protein. Sox9 plays both an essential role in
support the hypothesis that there is a higher rate of DSD the specification and differentiation of mesenchymal cells
in societies with a higher rate of consanguinity. The inci- toward the chondrogenic lineage through transcriptional
dence of ambiguous genitalia in Saudi Arabia has been modulation of Col2a1, the major matrix protein of the
estimated at 1:2,500 live births, whilst in Egypt it has been mature cartilage as well as establishing Sertoli cell iden-
estimated at 1:3,000 live births [Abdullah et al., 1991; Ma- tity in the developing testis immediately following the
zen et al., 2008], which is higher than the reported fre- expression of SRY. Many mutations have been reported
quency of 1:4,500–1:5,500 in European countries. Anoth- in the SOX9 gene and upstream and downstream flank-
er hindrance in defining the prevalence of DSD is the lack ing regions associated with campomelic dysplasia (CD).
of an accurate or even any diagnosis in many cases. In the There is a variable severity of testicular dysgenesis in
German study almost half of the children did not have a about 75% of affected XY individuals [Foster et al., 1994].
definitive diagnosis by the age of 6 months [Thyen et al., However, recently 2 patients with male external genita-
2006]. Excluding cases where the biochemical profile in- lia, unpalpable testis, and either hypospadias or micro-
dicates a specific error in steroidogenesis, it has been es- penis have been reported who carried p.Arg394Gly and
timated that a specific molecular diagnosis is obtained in p.Arg437Cys heterozygous missense mutations [Katoh-
about 20% of cases of DSD and that only 50% of 46,XY Fukui et al., 2015]. Neither of the boys had signs of CD,
children with DSD will receive a definitive clinical diag- although one boy carrying the p.Arg394Gly mutation was
nosis [Lee et al., 2006]. The detailed genetic analyses of reported as having spina bifida. Both of these mutations
individuals with DSD have been a powerful tool in the are located in the C-terminal PQS-rich domain of SOX9
identification of genes involved in sex determination and that is involved in protein-protein interactions with fac-
therefore DSD. tors such as CREB-binding protein and p300, both of
which positively regulate gene expression. The boy carry-
ing the p.Arg437Cys mutation presented with testicular
Gene Mutations and 46,XY Gonadal Dysgenesis regression sequence, reinforcing the hypothesis that both
gonadal dysgenesis and testicular regression sequence are
SRY and SOX9 part of the same phenotypic continuum.
Approximately 15% of 46,XY CGD patients carry mu- Besides the point mutations, rearrangements involv-
tations in the testis-determining gene SRY, with the ma- ing the SOX9 locus are also known to result in non-syn-
jority of these mutations localized within the functional dromic 46,XY or 46,XX DSD. The developmental timing
DNA-binding HMG-domain [McElreavey and Fellous, and tissue-specific transcriptional regulation of SOX9 is
1999] of the protein. Both CGD and PGD are also associ- highly complex and involves multiple elements located in
ated with small interstitial deletions 5′ and 3′ to the SRY flanking regions of at least 1 Mb upstream and 1.6 Mb
gene, as well as within the SRY promoter itself [McEl- downstream. Upstream of SOX9, translocations and in-
reavey et al., 1992, 1996; Assumpção et al., 2005]. In con- version breakpoints associated with CD fall within 2 clus-
trast to the mouse, the human SRY protein is expressed ters located about 400 kb apart [Leipoldt et al., 2007].
in Sertoli cells and germ cells from the moment of testis Large (>1 Mb) duplications 5′ to SOX9 are associated
determination until adulthood. The role, if any, of SRY in with brachydactyly-anonychia (symmetric brachydactyly
germ cells is unknown since SRY mutations are associ- of the hands/feet, hyponychia or anonychia) [Kurth et al.,
ated with a failure of testis development. SRY mutations 2009]. Pierre Robin sequence is a craniofacial disorder
are usually de novo, but some are inherited from a fertile characterized by micrognathia, cleft palate, and macro-
father. Functional studies suggest that these inherited glossia that is associated with normal testis development
mutant proteins retain partial biological activity, and the and is caused by either a 75-kb deletion located 1.38 Mb

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 315


Humans DOI: 10.1159/000452637
upstream or a deletion located 1.56 Mb downstream of factors, whereas strong transcriptional repression re-
SOX9 [Benko et al., 2009]. In mice, a testis-specific en- quires sumoylation of the lysines Lys119 and Lys194. Su-
hancer element has been mapped to a 1.4-kb region moylation plays an important role in NR5A1 function. If
termed Tesco that is located 13 kb upstream of Sox9 [Seki- it is eliminated from the mouse Nr5a1 protein, the mu-
do and Lovell-Badge, 2008]. Both Sry and Nr5a1 bind to tant mice exhibit marked endocrine abnormalities and
the Tesco enhancer sequence in vivo, possibly through a changes in cell fate that reflect an inappropriate activation
direct physical interaction to upregulate Sox9 gene ex- of hedgehog signaling [Lee et al., 2011].
pression. Once Sox9 protein levels reach a critical thresh- In humans, mutations involving NR5A1 are associated
old, several positive regulatory loops are initiated for its with a wide range of reproductive anomalies including
maintenance, including autoregulation of its own expres- 46,XY gonadal dysgenesis with or without adrenal insuf-
sion and formation of feed-forward loops via Ffg9 or ficiency, ambiguous genitalia, hypospadias, micropenis,
Pgd2 signaling. Other cofactors are likely to be involved spermatogenic failure with normal genitalia, and primary
in this process but have not yet been fully characterized. ovarian insufficiency [reviewed by El-Khairi and Acher-
Mutations involving the human TESCO element have not mann, 2012]. Familial cases have been described where
been reported as a cause of DSD, however, rearrange- both 46,XY gonadal dysgenesis and 46,XX ovarian insuf-
ments involving another regulatory element, termed ficiency are present in the same family [Fabbri et al.,
RevSex, located 600 kb upstream of SOX9, are associated 2014]. In a study of 315 men with spermatogenic failure,
with both XY and XX DSD. Five cases of 46,XX testicular we identified heterozygous missense mutations in NR5A1
or ovotesticular DSD that carried duplications of this re- in 7 men with either azoospermia or severe oligozoosper-
gion and a familial case of 46,XY DSD that carried a dele- mia [Bashamboo et al., 2010]. Testis histology in one man
tion of the element have been reported [Benko et al., 2011; with azoospermia was suggestive of a mild form of tes-
Cox et al., 2011; Vetro et al., 2011; Hyon et al., 2015]. The ticular dysgenesis rather than Sertoli-cell-only syndrome,
minimal region associated with 46,XX-SRY negative DSD again reinforcing the idea that errors in testis determina-
has been narrowed down to a 40.7–41.9-kb element, tion can manifest as different human reproductive phe-
which contains 2 predicted enhancer motifs [Hyon et al., notypes. In all cases, the men carrying the NR5A1 muta-
2015]. There is also data suggesting that deletions of an tions had normal development of the external genitalia
immediately adjacent and non-overlapping region are as- [Bashamboo et al., 2010]. The observation that mutations
sociated with 46,XY gonadal dysgenesis [Kim et al., 2015]. involving a key gene in human sex determination, NR5A1,
In our experience, about 10% of cases of 46,XX TDSD/ are associated with either male or female infertility es-
OTDSD and 46,XY gonadal dysgenesis have rearrange- tablishes a link between human sex determination and
ments involving the RevSex locus [unpubl. data]. fertility.
The mechanism behind the phenotype variability as-
NR5A1 sociated with NR5A1 mutations is yet to be explained,
Approximately 15% of all cases of 46,XY DSD are including variability associated with the same NR5A1
caused by mutations involving the gene NR5A1. NR5A1 mutation. For example, male infertility, female infertility,
belongs to the subfamily of transcription factors known or 46,XY DSD are associated with the variants p.Gly123Ala
as nuclear receptor subfamily 5 (group A, member 1), and p.Pro129Leu. It is possible that some patients with
which is highly conserved in vertebrates [Morohashi et NR5A1 mutations carry novel or rare variants in other
al., 1992]. The protein consists of a DNA-binding motif genes involved in sexual development that may influence
composed of 2 zinc-chelating modules that coordinate the severity of the phenotype. We reported 2 individuals
the interaction between the receptor and hormone re- with the same missense mutation p.Arg313Cys in NR5A1
sponse element [El-Khairi and Achermann, 2012]. [Allali et al., 2011; Mazen et al., 2016]. In the first case it
NR5A1 binds DNA as a monomer, with DNA binding was associated with mild hypospadias [Allali et al., 2011],
stabilized via a 30 amino acid extension termed the A- but in the second case it was associated with 46,XY go-
box. The C-terminal ligand-binding domain (LBD) is re- nadal dysgenesis [Mazen et al., 2016], and in both cases
quired for maximal biological activity with co-activators the mutation was de novo. The more severe phenotype
such as NCOA1. Posttranslational modification plays an may be explained by digenic inheritance since the patient
important role in modulating NR5A1 activation and re- carried a missense mutation in the MAP3K1 gene (see
pressor functions. Phosphorylation of Ser203 within the below). In other cases of DSD carrying mutations in
LBD enhances the positive interaction of regulatory co- NR5A1 the severity of the phenotype may be a conse-

316 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
quence of the specific amino acid involved (see NR5A1 in 46,XY DSD patients who have no evidence of heart
p.R92W below). A homozygous p.R103Q NR5A1 muta- disease [unpubl. data].
tion was reported in a child with severe 46,XY DSD and FOG2 (also known as ZFPM2) is a zinc finger cofactor
absent spleen. The mutation decreased the ability of the that modulates the activity of GATA4 by binding to the
mutated NR5A1 protein to transactivate TLX1, a tran- N-terminal zinc finger [Zaytouni et al., 2011]. XY Fog2–/–
scription factor that is essential for spleen development mice fail to develop testis, and since the expression of key
[Zangen et al., 2014]. The R103Q mutation impaired ac- genes involved in testis determination such as Sry and
tivation of steroidogenic genes without affecting the syn- Sox9 is dramatically reduced, it suggests that Fog2 is in-
ergistic NR5A1/SRY co-activation of SOX9. volved in the early stages of testis determination. The
evidence that FOG2 may be involved specifically in hu-
GATA4 and the Cofactor FOG2 man testis determination was suggested by 2 cases of
GATA4 belong to a class of evolutionarily conserved 46,XY gonadal dysgenesis in association with apparent-
lineage-limited zinc finger transcription factors charac- ly balanced translocations that included the FOG2 lo-
terized by the presence of 2 conserved type IV zinc finger cus on chromosome 8 (t(8;10)(q23.1;q21.1) and t(8;18)
domains that participate in cell fate determination, pro- (q22;q21)) [Finelli et al., 2007; Tan et al., 2012]. In both
liferation, and maturation [Zaytouni et al., 2011]. Both cases other complex somatic anomalies were reported.
GATA4 and GATA6 are expressed in the somatic tissues Using exome sequencing, we identified 2 independent
of the embryonic testis [Ketola et al., 1999]. GATA4 co- cases, of 46,XY gonadal dysgenesis each with missense
operatively interacts with NR5A1 to regulate the expres- mutations in the FOG2 gene [Bashamboo et al., 2014].
sion of genes critical for testis determination and differ- There was no history of cardiac anomalies in either the
entiation [Viger et al., 2008]. Male mice lacking Gata4 patients or their families. Functional studies indicated
show partially descended small testis with irregular that the failure of testis development in these cases could
cords, are infertile, and lack expression of Dmrt1 during be explained by the impaired ability of the mutant FOG2
embryogenesis [Manuylov et al., 2011]. Gata4ki mice, proteins to interact with GATA4. These studies estab-
which carry a p.Val217Gly mutation in the N-terminal lished GATA4 and FOG2 mutations as causes of 46,XY
zinc finger domain of the protein that abrogates the DSD.
physical interaction of Gata4 with the cofactor Fog2,
present with severe testicular dysgenesis [Molkentin et CBX2
al., 1997; Crispino et al., 2001; Bouma et al., 2007]. Gata4 CBX2 encodes for chromobox homolog 2, a compo-
also plays a pivotal role in Leydig cell function, for ex- nent of the polycomb group (PcG) complex of regulatory
ample a Gata4/Mef2 complex regulates Star gene expres- proteins. In mammals, PcG proteins are associated with
sion in mouse Leydig cells [Bergeron et al., 2015; Daems 2 main families of complexes, referred to as polycomb re-
et al., 2015; Schrade et al., 2015]. Although mutations in pressive complex 1 (PRC1) and PRC2. These complexes
human GATA4 are associated with congenital heart catalyze mono-ubiquitination of histone H2A on lysine
anomalies, a proportion of XY males carrying deletions 119 and tri-methylation of histone H3 on lysine 27, re-
of 8p23.1 that includes the GATA4 gene have hypospa- spectively. Human CBX2 exists in 2 isoforms, a 532 ami-
dias and bilateral cryptorchidism [Wat et al., 2009]. We no acid isoform termed CBX2.1 and a second shorter 211
identified a familial case of 46,XY DSD and congenital amino acid isoform termed CBX2.2. Mice lacking Cbx2
heart disease that affected both 46,XX and 46,XY indi- display posterior transformation of the vertebral columns
viduals [Lourenço et al., 2011]. This family carried a het- and sternal ribs, failure of T cell expansion, and XY mice
erozygous missense mutation (p.Gly221Arg) located im- show male-to-female sex reversal whereas XX animals
mediately adjacent to the mouse p.Val217Gly Gata4ki have either absent or smaller ovaries [Katoh-Fukui et al.,
mutation in the N-terminal zinc finger domain [Louren- 1998].
ço et al., 2011]. In functional studies the p.Gly221Arg A single patient with 46,XY gonadal dysgenesis and
variant failed to bind to DNA, did not to transactivate the mutations in the human CBX2 gene has been reported.
AMH promoter in a transient gene activation assay, and This was a 46,XY girl who carried 2 independent muta-
lacked the ability to bind to its protein partner FOG2. tions in CBX2 – a paternally inherited p.Pro98Leu muta-
Although the incidence of GATA4 mutations in associa- tion and a maternally inherited p.Arg443Pro mutation
tion with DSD has yet to be established, we have identi- [Biason-Lauber et al., 2009]. Histology of the gonads at
fied other heterozygous missense mutations in GATA4 4.5 years revealed apparently normal ovaries. Although

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 317


Humans DOI: 10.1159/000452637
polycomb group proteins are traditionally regarded as [Deng et al., 2006]. The MAP3K1 protein contains an
transcriptional repressors, there is evidence that at least amino terminal plant homeodomain (PHD) motif that
in some cellular or promoter contexts CBX2 acts as a has E3 Ub ligase activity, a caspase-3 cleavage site, and a
transcriptional activator of NR5A1 and SRY expression conserved kinase domain. MAP3K1 is the only MAP3K
[Biason-Lauber et al., 2009]. In the patient described that has a PHD motif. MAP3K1 is activated in response
above, the presence of apparently normal ovaries suggests to a number of different stimuli. These include growth
that CBX2 actively represses fetal ovarian development in factors, hyperosmolarity, microtubule disruption, cell
an XY individual. Recently, several potential downstream shape disturbance, pro-inflammatory cytokines, and
targets of CBX2 that are relevant to testis determination many other physiological stresses [Yujiri et al., 1998]. In
have been reported using a DamID-NGS approach [Eid humans, mutations in MAP3K1 have been identified in
et al., 2015]. The gene targets of CBX2 include many fac- cases of 46,XY DSD [Pearlman et al., 2010; Loke et al.,
tors that are known or suspected to be involved in sex 2014]. The mechanism whereby MAP3K1 mutations
development including SOX9, MAMLD1, SOX3, FGFR2, cause a failure of testis determination is unclear. The mu-
ATRX, TEX10, EXO1, TBX2, TSPYL4, WTAP, and MTM1 tations are heterozygous and are either missense, splice
[Eid et al., 2015]. The exact role of CBX2 (and by implica- site, or in-frame deletions. A clearly disruptive mutation,
tion the PcG complex) in sex determination is unknown, such as nonsense or frameshift mutation, has not been
although both the human and mouse phenotypes suggest identified, and considering the fact that MAP3K1 has a
that it is acting at an early stage of gonad formation. widespread expression pattern these more severe muta-
tions may be embryonic lethal. Available data suggest that
MAP3K1 mutations observed in DSD cases may be subtle gain-of-
The mitogen-activated protein kinases (MAPKs) are function variants that result in the increased phosphory-
activated through an evolutionarily conserved three- lation of the downstream MAPK proteins p38 MAPK and
component signal transduction cascade composed of a ERK1/2 [Pearlman et al., 2010; Loke et al., 2014]. Patients
mitogen-activated protein kinase kinase kinase 1 carrying these mutations show no other apparent pheno-
(MAP3K1), a MAP2K and a MAPK. Historically, these typic anomalies other than 46,XY gonadal dysgenesis [Le
were considered to be cellular housekeeping factors, and Caignec et al., 2003; unpubl. data].
it was a surprise to find that mutations in at least some of Mice lacking Map3K1 are viable and fertile but with an
these factors could generate gonad-specific phenotypes. increased embryonic gonadal length [Warr et al., 2011].
In mice, XY embryos lacking functional Map3k4 on a XY Map3k1mPHD/+ mice that are heterozygous for an in-
predominantly C57BL/6J background exhibit embryonic active PHD motif have a significantly enlarged testes but
gonadal XY sex reversal associated with a failure to tran- with a reduced number of Leydig cells [Charlaftis et al.,
scriptionally upregulate Sry [Bogani et al., 2009]. Mice 2014]. Another Map3k1 mouse model, goya, exhibits a
lacking Gadd45g, which encodes a protein that interacts severe hearing loss [Parker et al., 2015]. This data suggest
with Map3k4, also show a lack of testis determination that that in testis determination either the MAP kinase signal-
is associated with a delay in Sry expression [Gierl et al., ing pathways in human or mouse have diverged or the
2012]. Furthermore, the absence of both the p38a and difference in phenotype is caused by an intrinsic differ-
p38b MAPK isoforms results in XY sex reversal associ- ence in the type of mutation. In our experience about 10%
ated with reduced Sry expression [Warr et al., 2012]. Re- of 46,XY DSD cases with gonadal dysgenesis carry rare or
cent data also suggest that Map2k6 is required for the nor- novel variants in the MAP3K1 gene that could potential-
mal spatiotemporal expression profile of Sry [Warr et al., ly contribute to the phenotype. Although the MAP kinase
2016]. Therefore, at least in mice, available data are con- signaling pathway involved in mouse testis determina-
sistent with a GADD45γ/MAP3K4/p38 pathway which is tion is becoming clearer, the MAP3K1 pathway or the
required for the appropriate timing of Sry expression in targets of MAP Kinase signaling in human testis deter-
sex determination. mination are unknown. The phenotype associated with
Nineteen MAP3Ks are present in mammals, though MAP3K1 mutations is 46,XY CGD, a phenotype that is
their precise biological roles are not fully understood. also associated with mutations in the SRY gene. This sug-
MAP3K1 (also known as MEKK1) plays a role in lympho- gests that MAP3K1 signaling, like the equivalent pathway
cyte differentiation and function [Suddason and Galla- in the mouse, is required for the early stages of testis de-
gher, 2016], vasculature remodeling [Li et al., 2005], car- termination in humans, too.
diogenesis [Minamino et al., 2002], as well as injury repair

318 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
DMRT1, an Evolutionary Conserved Sex-Determining deviation from the genome-wide average number of non-
Gene synonymous mutations found in genes with a similar
Deletions of terminal 9p are associated with monoso- amount of global mutational burden (http://genic-intol-
my 9p syndrome, which is characterized by intellectual erance.org/). Mendelian disease causing genes are less
disability together with a distinctive series of somatic tolerant to coding variations than other genes. Put sim-
anomalies, and in approximately 70% of 46,XY individu- ply, genes known to carry few common functional vari-
als anomalies of testis development are seen that range ants in healthy individuals may be considered to be more
from a completely female phenotype to a male phenotype likely to cause certain diseases, such as rare forms of DSD,
with hypospadias and/or cryptorchidism [Ottolenghi than genes known to carry many functional variants. The
and McElreavey, 2000]. Two DMRT genes, DMRT1 and intolerance score is based upon allele frequency obtained
DMRT3 (DMRTA3), which are orthologues of the dou- from whole exome sequence data within the NHLBI-
blesex (dsx) of Drosophila and mab-3 of Caenorhabditis ESP6500 data set. A good example is the gene SOX9,
elegans, are located within the minimal recurrently de- which has an RVIS score of 14.4% and a deficit in loss-of-
leted region [Raymond et al., 1998]. Dsx controls the ter- function variants (%ExAC_RVIS) among the 9.81% low-
minal switch of the pathway leading to sex fate choice in est of the human genome. Equivalent %ExAC_RVIS fig-
Drosophila, and mab-3 is necessary to confer male traits ures for NR5A1, MAP3K1 and CBX2 are 15.76, 6.58 and
in C. elegans. In mice, Dmrt1 is not required for testis de- 11.63%, respectively. However, DMRT1 has an RVIS
termination, however, its continuous expression in the score of 25.56% and a deficit in loss-of-function variants
adult testis is required to maintain organ identity, because among the 40% lowest of the human genome. This indi-
forced attenuation of Dmrt1 expression in adult testis re- cates that DMRT1 is more tolerant to genetic variation in
sults in transdifferentiation of the testis to an ovary [Mat- healthy individuals than the other genes mentioned
son et al., 2011]. Although deletions of 9p24 suggest that above. This suggests that for a DMRT1 mutation to be
DMRT1 hemizygosity is sufficient in some individuals to pathogenic (or penetrant) resulting in a failure of testis
lead to a failure of testicular development, these deletions determination, it may be required to show either domi-
usually remove other genes, including the evolutionary nant negative activity on the wild-type allele or alter the
related DMRT2 and DMRT3 genes [Ottolenghi and normal interactions of the protein.
McElreavey, 2000]. Formally, the phenotype could be due Gonadal anomalies may not be the only phenotype as-
to haploinsufficiency of 1, 2, or all 3 of these DMRT genes. sociated with mutations involving DMRT1. Recently, ge-
Evidence to indicate that the key player in human testis nome-wide association studies indicated a variant in the
determination is DMRT1 came through the identification putative promoter of DMRT1 for sex-specific asthma
of a de novo missense mutation in the functionally im- [Schieck et al., 2016]. The role, if any, of DMRT1 in the
portant DM-DNA-binding domain in a patient with regulation of allergic immune responses is unclear, but
46,XY CGD [Murphy et al., 2015]. There were no other the expression of DMRT1 was found to be higher in lung
somatic anomalies in this healthy girl. The histology of macrophages from men with various lung diseases
the gonad was similar to that of an SRY mutation and [Schieck et al., 2016].
showed no evidence of testicular material, suggesting that
the mutation was indeed impacting on primary testis de-
termination. In vitro studies indicated that the mutant 46,XX Testicular and Ovotesticular DSD
protein had reduced DNA affinity, altered sequence spec-
ificity and when mixed with wild-type protein, it altered SOX Gene Mutations
the stoichiometry of the wild-type protein [Murphy et al., In recent years it has become evident that the ectopic
2015]. This suggests that the lack of testis determination expression of HMG-box containing proteins in the uro-
seen in this patient is due to a combination of haploinsuf- genital ridge at the moment of sex determination may
ficiency and dominant negative activity. This observation result in testicular development in a chromosomal XX
may also explain, at least in part, the absence of mutations female. Although most cases of 46,XX TDSD/OTDSD are
that have been identified thus far in cases of 46,XY go- caused by the presence of the SRY gene, usually on the X
nadal dysgenesis. In a screen of over 100 cases of XY go- chromosome, the remaining cases stay unexplained. As
nadal dysgenesis, we have only identified a single muta- compared to 46,XY DSD, there are relatively fewer known
tion associated with the phenotype. The residual varia- causes of 46,XX DSD. In human, SOX3 loss-of-function
tion intolerance score (RVIS) ranks human genes by their mutations are associated with mental retardation and

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 319


Humans DOI: 10.1159/000452637
growth hormone deficiency [Laumonnier et al., 2002]. sociated with exceptionally rare syndromic forms of
However, three 46,XX SRY-negative testicular DSD pa- 46,XX testicular/ovotesticular DSD. Human homozy-
tients have been reported who carry rearrangements at gous RSPO1 mutations are associated with a rare reces-
the SOX3 locus on the X chromosome. One patient car- sive syndrome, which is characterized by XX testicular
ried 2 microduplications, one of ∼123 kb that spanned DSD, palmoplantar hyperkeratosis, and predisposition to
the entire SOX3 gene and another of 85 kb that was lo- squamous cell carcinoma of the skin [Parma et al., 2006].
cated 350 kb proximal to SOX3. A second patient carried Mutations involving RSPO1 have not been reported in
a single 343-kb microdeletion immediately upstream of non-syndromic cases of testicular and ovotesticular DSD
SOX3, and a third XX male with multiple congenital [unpubl. data].
anomalies carried a 6-Mb duplication including SOX3 The absence of Wnt4 in XX mice results in a partial
and at least 18 other genes [Sutton et al., 2011]. 46,XX masculinization of the gonad including the differentia-
testicular DSD has also been reported in association with tion of some Leydig-like cells. In human, 4 dominant
a 774-kb insertion translocation from chromosome 1 into heterozygous missense mutations in WNT4 have been
a palindromic sequence 82 kb distal to SOX3 [Haines et reported in 46,XX women with various degrees of viril-
al., 2015]. Three further duplications of the SOX3 gene ization including androgen excess and abnormal devel-
have been reported in XX males [Moalem et al., 2012; opment of Müllerian ducts [Baison-Lauber et al., 2004,
Vetro et al., 2015; Grinspon et al., 2016]. 2007; Philibert et al., 2008, 2011]. A single homozygous
Complete or partial duplications of chromosome 22 in WNT4 mutation was reported in a consanguineous fam-
46,XX-SRY negative individuals are associated with vari- ily with an embryonic lethal syndrome of 46,XX testicular
ous degrees of masculinization [Nicholl et al., 1994; Aleck DSD and dysgenesis of kidneys, adrenals, and lungs
et al., 1999; Seeherunvong et al., 2004]. Further delimita- (SERKAL syndrome; SEx Reversion, Kidneys, Adrenal
tion of the minimal region was demonstrated by a de and Lung dysgenesis) [Mandel et al., 2008].
novo duplication of 22q11.2q13 in a 46,XX SRY-negative
male with mild hypospadias, dysmorphic features, and NR5A1 p.Arg92Trp and 46,XX DSD
hypotonia [Polanco et al., 2010]. Human SOX10 maps to Primary ovarian insufficiency, also termed premature
22q13.1 and may be responsible for the phenotype. Loss- ovarian failure, is defined by the arrest of normal ovarian
of-function heterozygous mutations in SOX10 are as- function before the age of 40 years and includes prema-
sociated with Waardenburg-Shah and Waardenburg- ture menopause, primary and secondary amenorrhea as
Hirschsprung disease [Pingault et al., 1998; Touraine et well as ovarian dysgenesis. In 2007, when we were ana-
al., 2000], however, in the mouse, transgenic expression lyzing cases of 46,XX primary ovarian insufficiency
of Sox10 in the gonads of XX mice results in testis forma- for mutations in the NR5A1 gene, a patient (sporadic case
tion [Polanco et al., 2010]. 2; Lourenco et al. [2009]) was clinically investigated at 4
months of age because of a hypertrophy of the clitoris.
RSPO1/WNT4/β-Catenin Signaling The girl had high levels of FSH indicating ovarian insuf-
Little is known about the genetic pathway(s) involved ficiency and carried an NR5A1 mutation. The clitoral hy-
in human ovary development. In XX individuals, activa- pertrophy suggested that the girl had been exposed to an-
tion of the β-catenin signaling pathway by the proteins drogens in utero. Therefore, we decided to screen idio-
RSPO1 and WNT4 is necessary for granulosa cell differ- pathic cases of 46,XX TDSD/OTDSD for mutations in the
entiation leading to ovarian development. Stabilization of NR5A1 gene. We rapidly discovered a small family with
β-catenin by the RSPO1/WNT4 pathway results in tran- 2 virilized sibs who carried a p.Arg92Trp mutation. The
scription of its target genes. The mechanism by which unaffected mother also carried this mutation. At the time,
RSPO1 stimulates WNT4 in the developing ovary is un- this observation was interesting but could simply have
known. In general, RSPOs stimulate WNT signaling by been explained as a chance finding – a rare genetic variant
binding to the leucine-rich repeat-containing G protein- unrelated to the phenotype. This view was supported by
coupled receptors LGR4, LGR5, and LGR6 [Wang et al., our screen of further 40 cases of 46,XX DSD that did not
2013]. RSPOs can also bind to 2 negative feedback regula- reveal any other NR5A1 mutations. In 2015, we were con-
tors of the WNT signaling pathway, the RING-type E3 tacted by different groups who had also identified the
ubiquitin ligases ZNRF3 or RNF43, leading to their clear- same NR5A1 p.Arg92Trp mutation in association with
ance and resulting in enhanced WNT signaling [Jiang et 46,XX testicular DSD [Bashamboo et al., 2016]. Two fam-
al., 2015]. Mutations involving RSPO1 and WNT4 are as- ilies had a single affected child who carried a de novo

320 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
Fig. 1. NR5A1 may regulate anti-testis gene expression in the ova- gene expression leading to a lack of testis formation. In a 46,XX
ry. In a 46,XY individual, NR5A1 synergizes with SRY to upregu- child with TDSD/OTDSD (shown on the right), the same mutant
late male-specific gene expression (e.g., SOX9) leading to testis for- shows reduced ability to synergize with β-catenin to upregulate the
mation. In 46,XX individuals, NR5A1 synergizes with β-catenin to expression of anti-testis genes. As a consequence of this lack of re-
upregulate the expression of anti-testis genes (e.g., DAX-1/NR0B1) pression, the expression of pro-testis genes (e.g., SOX9) leads to
and possibly pro-ovarian genes. In the 46,XY DSD case, the testis formation.
p.Arg92Trp mutant shows a reduced ability to upregulate SOX9

NR5A1 p.Arg92Trp mutation, and this formally excluded of pathways that oppose testis development and maintain
a founder mutation. A fourth family was identified with ovarian integrity. Transient transfection assays demon-
2 affected sibs, one with 46,XY gonadal dysgenesis and strated that the p.Arg92Trp NR5A1 mutant had reduced
raised as a girl and the other a boy with 46,XX testicular activation of several minimal promoters involved in testis
DSD. They both carried the mutation. A number of mu- development as well as the Sox9 Tesco enhancer. This is
tations have been described in NR5A1 in 46,XX indi- consistent with a 46,XY PGD phenotype, but it does not
viduals in association with ovarian insufficiency, but we explain testis formation in an XX background. We then
are unaware of any other amino acid changes in NR5A1 ex- assayed the ability of the 92Trp and 92Gln mutations to
cept p.Arg92Trp that is associated with virilization in influence the pro-ovary canonical WNT signaling path-
46,XX individuals. Interestingly, a homozygous p.Arg92Gln way. β-Catenin can interact functionally with NR5A1 to
change has been observed in a 46,XX girl with no evi- modulate target gene expression [Gummow et al., 2003;
dence of virilization [Guran et al., 2015]. This suggests Hossain and Saunders, 2003; Jordan et al., 2003; Kennell
that the p.Arg92Trp mutation specifically results in testis et al., 2003; Mizusaki et al., 2003; Parakh et al., 2006; Salis-
formation in a chromosomal female background (fig. 1). bury et al., 2007; Ehrlund et al., 2012]. Specifically, the
The mutation involves a highly conserved amino acid res- Nr5a1 and β-catenin proteins physically interact to up-
idue located in the A-box motif of the protein. The A-box regulate the expression of the Nr0b1 (Dax-1) gene on the
consists of a 30 amino acid basic region carboxyl-terminal X chromosome [Mizusaki et al., 2003]. In 46,XY individ-
to the DNA binding domain. NR5A1 binds to DNA as a uals, 2 copies of NR0B1 result in a failure of testis deter-
monomer with the A-box recognizing DNA sequences 5′ mination and are associated with 46,XY gonadal dysgen-
to the NR5A1 consensus motif in the minor groove. The esis, indicating that NR0B1 is an anti-testis gene [Mus-
p.Arg92Trp mutation is predicted to disrupt DNA bind- catelli et al., 1994]. In contrast to the p.Arg92Gln mutant,
ing and this is what we observed using the consensus we found that the p.Arg92Trp NR5A1 mutant showed
binding motif CCAAGGTCA as a target. In contrast the loss of synergy with β-catenin to activate target gene ex-
p.Arg92Gln mutant has essentially wild-type binding ac- pression. The phenotype in the 46,XX children with the
tivity. However, this difference in DNA binding activity p.Arg92Trp variant may be due to altered regulation of
cannot explain why testis determination is occurring due the expression of pro-ovarian genes that normally sup-
to the 92Trp substitution. The p.Arg92Trp mutation press testis development (fig.  2). In humans, NR5A1 is
could be associated either with inappropriate activation expressed in the granulosa cells of the early developing
of testis-specific pathways in the ovary or with disruption ovary, whereas in the mouse the expression of Nr5a1 in

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 321


Humans DOI: 10.1159/000452637
Schematic diagram of mammalian somatic sex determination

MAP kinases
Genital ridge
GATA4/FOG2/WT1 ?
SRY
FGF9, DMRT1 NR5A1 DHH
PTGDS
?
WNT4 -catenin Foetal
SOX9 Sox8/SOX9 Leydig Cell
RSPO1
NR5A1,GATA4
? NR5A1
WT1, SOX8
NR5A1
AMH steroidogenesis

NR5A1

steroidogenesis DMRT1
DMRT1?
Adult
FOXL2 SOX9 FOXL2 SOX9

TIME
OVARY TESTIS

Fig. 2. The molecular and genetic events in mammalian sex deter- expression and formation of feed-forward loops via FGF9 or PGD2
mination and differentiation. In the XY gonad the activation of signaling. At later stages, Foxl2 may repress Sox9 expression to
SRY expression, possibly initiated by CBX2/WT1/GATA4/FOG2/ maintain ovarian identity [Uhlenhaut et al., 2009]. In the testis,
NR5A1, leads to the upregulation of Sox9 expression via a synergy Sox9, together with other Sox proteins including Sox8, promotes
with Nr5a1 at a Sox9 enhancer such as Tesco [Sekido and Lovell- the testis pathway by for example stimulating Amh expression, and
Badge, 2008]. In the XX gonad the supporting cell precursors ac- it also probably represses the ovarian genes Wnt4 and Foxl2
cumulate β-catenin in response to RSPO1/WNT4 signaling, which [Uhlenhaut et al., 2009]. DMRT1 controls sex determination in
either directly or indirectly represses SOX9 expression, perhaps at some species of fish and may be the master sex-determining switch
least in human, by interacting with NR5A1 [Bashamboo et al., in birds. In mice it is not involved in male primary sex determina-
2016]. Once Sox9 levels reach a critical threshold, several positive tion, but there is evidence to indicate that it is important in human
regulatory loops are initiated, including autoregulation of its own testis determination [Murphy et al., 2015].

the embryonic gonad is sexually dimorphic. It is continu- Conclusions


ously expressed in the mouse embryonic testis, but Nr5a1
transcripts are absent during the period of ovarian forma- The last few years have seen a considerable number of
tion between E13.5–E16.5 [Ikeda et al., 1994]. Therefore new genes involved in human sex determination that
in this case, the mouse model may not be informative for when mutated cause non-syndromic DSD. A failure of
elucidation of the mechanism. This further reiterates the testis determination, 46,XY gonadal dysgenesis is mainly
differences between the molecular mechanisms involved associated with point mutations in coding sequences (e.g.
in gonad determination in mouse and human and em- NR5A1, MAP3K1, GATA4, FOG2), whereas gene muta-
phasizes the need to analyze human cases of DSD to es- tions leading to testis formation in a chromosomal XX
tablish novel causes of 46,XX and 46,XY DSD. female background are mainly associated with dysregula-
tion of the expression of SOX genes (e.g. SOX9, SOX3,

322 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
SOX10). In the latter, point mutations in coding genes are Acknowledgements
usually associated with syndromic forms of DSD involv-
A.B. is funded in part by the program Actions Concertees Inter-
ing the RSPO/WNT signaling pathway. Overall, the ge- pasteuriennes (ACIP). A.B. and K.McE. are funded by a research
netic etiology in the majority of cases of these extreme grant from the EuroDSD in the European Community’s Seventh
forms of DSD is unknown and reflects our poor knowl- Framework Programme FP7/2007-2013 under grant agreement No.
edge of the genetic pathways that are involved. High 201444 as well as grant No. 295097 entitled GM_NCD_in_Co – Re-
throughput sequencing will reveal rare genetic mutations inforcing IPT capacities in Genomic Medicine, Non Communicable
Diseases Investigation and international cooperation as part of the
that are responsible for these phenotypes; however, if we EU call FP7-INCO-2011-6. The work is also funded by a Franco-
are to fully establish the causality of these mutations and Egyptian AIRD-STDF grant and the Agence Nationale de la Recher-
understand the mechanisms, a series of complimentary che (Laboratoire d’Excellence Revive).
combinatorial studies using different model systems are
required.
Disclosure Statement

The authors have no conflicts of interest to declare.

References
Abdullah MA, Katugampola M, al-Habib S, al- rian-duct regression and virilization in a Charlaftis N, Suddason T, Wu X, Anwar S, Karin
Jurayyan N, al-Samarrai A, et al: Ambiguous 46,XX woman. N Engl J Med 351: 792–798 M, et al: The MEKK1 PHD ubiquitinates
genitalia: medical, socio-cultural and reli- (2004). TAB1 to activate MAPKs in response to cyto-
gious factors affecting management in Saudi Biason-Lauber A, De Filippo G, Konrad D, Scar- kines. EMBO J 33:2581–2596 (2014).
Arabia. Ann Trop Paediatr 11: 343–348 ano G, Nazzaro A, et al: WNT4 deficiency – a Cox JJ, Willatt L, Homfray T, Woods CG: A SOX9
(1991). clinical phenotype distinct from the clas- duplication and familial 46,XX developmen-
Aleck KA, Argueso L, Stone J, Hackel JG, Erick- sic Mayer-Rokitansky-Kuster-Hauser syn- tal testicular disorder. N Engl J Med 364: 91–
son RP: True hermaphroditism with partial drome: a case report. Hum Reprod 22: 224– 93 (2011).
duplication of chromosome 22 and without 229 (2007). Crispino JD, Lodish MB, Thurberg BL, Litovsky
SRY. Am J Med Genet 85:2–4 (1999). Biason-Lauber A, Konrad D, Meyer M, DeBeau- SH, Collins T, et al: Proper coronary vascular
Allali S, Muller JB, Brauner R, Lourenço D, Boud- fort C, Schoenle EJ: Ovaries and female phe- development and heart morphogenesis de-
jenah R, et al: Mutation analysis of NR5A1 en- notype in a girl with 46,XY karyotype and pend on interaction of GATA-4 with FOG co-
coding steroidogenic factor 1 in 77 patients mutations in the CBX2 gene. Am J Hum Ge- factors. Genes Dev 15:839–844 (2001).
with 46,XY disorders of sex development net 84:658–663 (2009). Daems C, Di-Luoffo M, Paradis É, Tremblay JJ:
(DSD) including hypospadias. PLoS One 6: Benko S, Fantes JA, Amiel J, Kleinjan DJ, Thomas MEF2 cooperates with Forskolin/cAMP and
e24117 (2011). S, et al: Highly conserved non-coding ele- GATA4 to regulate Star gene expression in
Arlt W, Krone N: Adult consequences of congen- ments on either side of SOX9 associated with mouse MA-10 leydig cells. Endocrinology
ital adrenal hyperplasia. Horm Res 68: 158– Pierre Robin sequence. Nat Genet 41:359–364 156:2693–2703 (2015).
164 (2007). (2009). de la Chapelle A: Analytic review: nature and ori-
Assumpção JG, Ferraz LF, Benedetti CE, Maciel- Benko S, Gordon CT, Mallet D, Sreenivasan R, gin of males with XX sex chromosomes. Am J
Guerra AT, Guerra G Jr, et al: A naturally oc- Thauvin-Robinet C, et al: Disruption of a long Hum Genet 24:71–105 (1972).
curring deletion in the SRY promoter region distance regulatory region upstream of SOX9 Deng M, Chen WL, Takatori A, Peng Z, Zhang L,
affecting the Sp1 binding site is associated in isolated disorders of sex development. J et al: A role for the mitogen-activated protein
with sex reversal. J Endocrinol Invest 28:651– Med Genet 48:825–830 (2011). kinase kinase kinase 1 in epithelial wound
656 (2005). Bergeron F, Nadeau G, Viger RS: GATA4 knock- healing. Mol Biol Cell 17:3446–3455 (2006).
Bashamboo A, Ferraz-de-Souza B, Lourenço D, down in MA-10 Leydig cells identifies multi- Ehrlund A, Jonsson P, Vedin LL, Williams C,
Lin L, Sebire NJ, et al: Human male infertility ple target genes in the steroidogenic pathway. Gustafsson JÅ, et al: Knockdown of SF-1 and
associated with mutations in NR5A1 encod- Reproduction 149:245–257 (2015). RNF31 affects components of steroidogene-
ing steroidogenic factor 1. Am J Hum Genet Bogani D, Siggers P, Brixey R, Warr N, Beddow S, sis, TGFβ, and Wnt/β-catenin signaling in ad-
87:505–512 (2010). et al: Loss of mitogen-activated protein kinase renocortical carcinoma cells. PLoS One 7:
Bashamboo A, Brauner R, Bignon-Topalovic J, kinase kinase 4 (MAP3K4) reveals a require- e32080 (2012).
Lortat-Jacob S, Karageorgou V, et al: Muta- ment for MAPK signalling in mouse sex de- Eid W, Opitz L, Biason-Lauber A: Genome-wide
tions in the FOG2/ZFPM2 gene are associated termination. PLoS Biol 7:e1000196 (2009). identification of CBX2 targets: insights in the
with anomalies of human testis determina- Bouma GJ, Washburn LL, Albrecht KH, Eicher human sex development network. Mol Endo-
tion. Hum Mol Genet 23:3657–3665 (2014). EM: Correct dosage of Fog2 and Gata4 tran- crinol 29:247–257 (2015).
Bashamboo A, Donohoue PA, Vilain E, Rojo S, scription factors is critical for fetal testis de- El-Khairi R, Achermann JC: Steroidogenic fac-
Calvel P, et al: A recurrent p.Arg92Trp variant velopment in mice. Proc Natl Acad Sci USA tor-1 and human disease. Semin Reprod Med
in steroidogenic factor-1 (NR5A1) can act as 104:14994–14999 (2007). 30:374–381 (2012).
a molecular switch in human sex develop- Brown S, Yu C, Lanzano P, Heller D, Thomas L, Ergun-Longmire B, Vinci G, Alonso L, Matthew
ment. Hum Mol Genet doi: 10.1093/hmg/ et al: A de novo mutation (Gln2Stop) at the 5′ S, Tansil S: Clinical, hormonal and cytogenet-
ddw186 (2016). end of the SRY gene leads to sex reversal with ic evaluation of 46,XX males and review of the
Biason-Lauber A, Konrad D, Navratil F, Schoenle partial ovarian function. Am J Hum Genet 62: literature. J Pediatr Endocrinol Metab 18:
EJ: A WNT4 mutation associated with Mülle- 189–192 (1998). 739–748 (2005).

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 323


Humans DOI: 10.1159/000452637
Fabbri HC, de Andrade JG, Soardi FC, de Calais factor 1/beta-catenin synergy. Proc Natl Acad Li Y, Minamino T, Tsukamoto O, Yujiri T, Shin-
FL, Petroli RJ: The novel p.Cys65Tyr muta- Sci USA 100:10866–10871 (2003). tani Y, et al: Ablation of MEK kinase 1 sup-
tion in NR5A1 gene in three 46,XY siblings Katoh-Fukui Y, Tsuchiya R, Shiroishi T, Naka- presses intimal hyperplasia by impairing
with normal testosterone levels and their hara Y, Hashimoto N, et al: Male-to-female smooth muscle cell migration and urokinase
mother with primary ovarian insufficiency. sex reversal in M33 mutant mice. Nature 393: plasminogen activator expression in a mouse
BMC Med Genet 15:17 (2014). 688–692 (1998). blood-flow cessation model. Circulation 111:
Finelli P, Pincelli AI, Russo S, Bonati MT, Recal- Katoh-Fukui Y, Igarashi M, Nagasaki K, Horika- 1672–1678 (2005).
cati MP: Disruption of friend of GATA 2 gene wa R, Nagai T, et al: Testicular dysgenesis/re- Loke J, Pearlman A, Radi O, Zuffardi O, Giussani
(FOG-2) by a de novo t(8; 10) chromosomal gression without campomelic dysplasia in U, et al: Mutations in MAP3K1 tilt the balance
translocation is associated with heart defects patients carrying missense mutations and from SOX9/FGF9 to WNT/β-catenin signal-
and gonadal dysgenesis. Clin Genet 71: 195– upstream deletion of SOX9. Mol Genet Ge- ing. Hum Mol Genet 23:1073–1083 (2014).
204 (2007). nomic Med 3:550–557 (2015). Lourenço D, Brauner R, Rybczynska M, Nihoul-
Foster JW, Dominguez-Steglich MA, Guioli S, Kennell JA, O’Leary EE, Gummow BM, Hammer Fékété C, McElreavey K, et al: Loss-of-func-
Kwok C, Weller PA, et al: Campomelic dys- GD, MacDougald OA: T-cell factor 4N (TCF- tion mutation in GATA4 causes anomalies of
plasia and autosomal sex reversal caused by 4N), a novel isoform of mouse TCF-4, syner- human testicular development. Proc Natl
mutations in an SRY-related gene. Nature gizes with β-catenin to coactivate C/EBPα and Acad Sci USA 108:1597–1602 (2011).
372:525–530 (1994). steroidogenic factor 1 transcription factors. Mandel H, Shemer R, Borochowitz ZU, Okopnik
Gierl MS, Gruhn WH, von Seggern A, Maltry N, Mol Cell Biol 23:5366–5375 (2003). M, Knopf C, et al: SERKAL syndrome: an au-
Niehrs C: GADD45G functions in male sex Ketola I, Rahman N, Toppari J, Bielinska M, Por- tosomal-recessive disorder caused by a loss-
determination by promoting p38 signaling ter-Tinge SB, et al: Expression and regulation of-function mutation in WNT4. Am J Hum
and Sry expression. Dev Cell 23: 1032–1042 of transcription factors GATA-4 and GATA- Genet 82:39–47 (2008).
(2012). 6 in developing mouse testis. Endocrinology Manuylov NL, Zhou B, Ma Q, Fox SC, Pu WT, et
Grinspon RP, Nevado J, Mori Alvarez ML, Del 140:1470–1480 (1999). al: Conditional ablation of Gata4 and Fog2
Rey G, Castera R, et al: 46,XX ovotesticular Kim GJ, Sock E, Buchberger A, Just W, Denzer F, genes in mice reveals their distinct roles in
DSD associated with a SOX3 gene duplication et al: Copy number variation of two separate mammalian sexual differentiation. Dev Biol
in a SRY-negative boy. Clin Endocrinol (Oxf) regulatory regions upstream of SOX9 causes 353:229–241 (2011).
85:673–675 (2016). isolated 46,XY or 46,XX disorder of sex devel- Marcantonio SM, Fechner PY, Migeon CJ, Perl-
Gummow BM, Winnay JN, Hammer GD: Con- opment. J Med Genet 52:240–247 (2015). man EJ, Berkovitz GD: Embryonic testicular
vergence of Wnt signaling and steroidogenic Kurth I, Klopocki E, Stricker S, van Oosterwijk J, regression sequence: a part of the clinical
factor-1 (SF-1) on transcription of the rat in- Vanek S, et al: Duplications of noncoding ele- spectrum of 46,XY gonadal dysgenesis. Am J
hibin alpha gene. J Biol Chem 278: 26572– ments 5′ of SOX9 are associated with brachy- Med Genet 49:1–5 (1994).
26579 (2003). dactyly-anonychia. Nat Genet 41: 862–863 Marino M, Moriondo V, Vighi E, Pignatti E, Si-
Guran T, Buonocore F, Saka N, Ozbek MN, Ay- (2009). moni M: Central hypogonadotropic hypogo-
can Z, et al: Rare causes of primary adrenal Laumonnier F, Ronce N, Hamel BC, Thomas P, nadism: genetic complexity of a complex dis-
insufficiency: genetic and clinical character- Lespinasse J, et al: Transcription factor SOX3 ease. Int J Endocrinol 2014:649154 (2014).
ization of a large nationwide cohort. J Clin is involved in X-linked mental retardation Matson CK, Murphy MW, Sarver AL, Griswold
Endocrinol Metab 101:284–292 (2016). with growth hormone deficiency. Am J Hum MD, Bardwell VJ, et al: DMRT1 prevents fe-
Haines B, Hughes J, Corbett M, Shaw M, Innes J, Genet 71:1450–1455 (2002). male reprogramming in the postnatal mam-
et al: Interchromosomal insertional translo- Le Caignec C, Baron S, McElreavey K, Joubert M, malian testis. Nature 476:101–104 (2011).
cation at Xq26.3 alters SOX3 expression in an Rival JM, et al: 46,XY gonadal dysgenesis: ev- Mazen I, Hiort O, Bassiouny R, El Gammal M:
individual with XX male sex reversal. J Clin idence for autosomal dominant transmission Differential diagnosis of disorders of sex de-
Endocrinol Metab 100:815–820 (2015). in a large kindred. Am J Med Genet A velopment in Egypt. Horm Res 70: 118–123
Hossain A, Saunders GF: Synergistic cooperation 116A:37–43 (2003). (2008).
between the beta-catenin signaling pathway Lee FY, Faivre EJ, Suzawa M, Lontok E, Ebert D, Mazen I, Abdel-Hamid M, Mekkawy M, Bignon-
and steroidogenic factor 1 in the activation of et al: Eliminating SF-1 (NR5A1) sumoylation Topalovic J, Boudjenah R, et al: Identification
the Mullerian inhibiting substance type II re- in vivo results in ectopic hedgehog signaling of NR5A1 mutations and possible digenic in-
ceptor. J Biol Chem 278:26511–26516 (2003). and disruption of endocrine development. heritance in 46,XY gonadal dysgenesis. Sex
Hyon C, Chantot-Bastaraud S, Harbuz R, Bhouri Dev Cell 21:315–327 (2011). Dev 10:147–151 (2016).
R, Perrot N, et al: Refining the regulatory re- Lee PA, Houk CP, Ahmed SF, Hughes IA: Inter- McElreavey K, Fellous M: Sex determination and
gion upstream of SOX9 associated with 46,XX national Consensus Conference on Intersex the Y chromosome. Am J Med Genet 89:176–
testicular disorders of sex development organized by the Lawson Wilkins Pediatric 185 (1999).
(DSD). Am J Med Genet A 167: 1851–1858 Endocrine Society and the European Society McElreavy K, Vilain E, Abbas N, Costa JM, Sou-
(2015). for Paediatric Endocrinology. Consensus leyreau N et al: XY sex reversal associated with
Ikeda Y, Shen WH, Ingraham HA, Parker KL: De- statement on management of intersex disor- a deletion 5′ to the SRY ‘HMG box’ in the tes-
velopmental expression of mouse steroido- ders. International Consensus Conference on tis-determining region. Proc Natl Acad Sci
genic factor-1, an essential regulator of the Intersex. Pediatrics 118:488–500 (2006). USA 89:11016–11020 (1992).
steroid hydroxylases. Mol Endocrinol 8:654– Lee PA, Nordenström A, Houk CP, Ahmed SF, McElreavey K, Vilain E, Barbaux S, Fuqua JS,
662 (1994). Auchus R, et al: Global disorders of sex devel- Fechner PY, et al: Loss of sequences 3′ to the
Jiang X, Charlat O, Zamponi R, Yang Y, Cong F: opment update since 2006: perceptions, ap- testis-determining gene, SRY, including the Y
Dishevelled promotes Wnt receptor degrada- proach and care. Horm Res Paediatr 85:158– pseudoautosomal boundary associated with
tion through recruitment of ZNRF3/RNF43 180 (2016). partial testicular determination. Proc Natl
E3 ubiquitin ligases. Mol Cell 58: 522–533 Leipoldt M, Erdel M, Bien-Willner GA, Smyk M, Acad Sci USA 93:8590–8594 (1996).
(2015). Theurl M, et al: Two novel translocation Minamino T, Yujiri T, Terada N, Taffet GE, Mi-
Jordan BK, Shen JH, Olaso R, Ingraham HA, breakpoints upstream of SOX9 define borders chael LH, et al: MEKK1 is essential for car-
Vilain E: Wnt4 overexpression disrupts nor- of the proximal and distal breakpoint cluster diac hypertrophy and dysfunction induced
mal testicular vasculature and inhibits testos- region in campomelic dysplasia. Clin Genet by Gq. Proc Natl Acad Sci USA 99:3866–3871
terone synthesis by repressing steroidogenic 71:67–75 (2007). (2002).

324 Sex Dev 2016;10:313–325 Bashamboo/McElreavey


DOI: 10.1159/000452637
Mizusaki H, Kawabe K, Mukai T, Ariyoshi E, amenorrhea and Müllerian duct abnormali- Thyen U, Lanz K, Holterhus PM, Hiort O: Epide-
Kasahara M, et al: Dax-1 (dosage-sensitive sex ties: a French collaborative study. J Clin En- miology and initial management of ambigu-
reversal-adrenal hypoplasia congenita critical docrinol Metab 93:895–900 (2008). ous genitalia at birth in Germany. Horm Res
region on the X chromosome, gene 1) gene Philibert P, Biason-Lauber A, Gueorguieva I, 66:195–203 (2006).
transcription is regulated by wnt4 in the fe- Stuckens C, Pienkowski C, et al: Molecular Touraine RL, Attié-Bitach T, Manceau E, Korsch
male developing gonad. Mol Endocrinol 17: analysis of WNT4 gene in four adolescent E, Sarda P, et al: Neurological phenotype in
507–519 (2003). girls with Müllerian duct abnormality and Waardenburg syndrome type 4 correlates
Moalem S, Babul-Hirji R, Stavropolous DJ, Wher- hyperandrogenism (atypical Mayer-Rokitan- with novel SOX10 truncating mutations and
rett D, Bägli DJ, et al: XX male sex reversal sky-Küster-Hauser syndrome). Fertil Steril expression in developing brain. Am J Hum
with genital abnormalities associated with a 95:2683–2686 (2011). Genet 66:1496–1503 (2000).
de novo SOX3 gene duplication. Am J Med Phillips NB, Racca J, Chen YS, Singh R, Jancso- Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T,
Genet A 158A:1759–1764 (2012). Radek A, et al: Mammalian testis-determin- Sekido R, et al: Somatic sex reprogramming
Molkentin JD, Lin Q, Duncan SA, Olson EN: Re- ing factor SRY and the enigma of inherited of adult ovaries to testes by FOXL2 ablation.
quirement of the transcription factor GATA4 human sex reversal: frustrated induced fit in Cell 139:1130–1142 (2009).
for heart tube formation and ventral morpho- a bent protein-DNA complex. J Biol Chem Vetro A, Ciccone R, Giorda R, Patricelli MG, Del-
genesis. Genes Dev 11:1061–1072 (1997). 286:36787–36807 (2011). la Mina E, et al: XX males SRY negative: a con-
Morohashi K, Honda S, Inomata Y, Handa H, Pingault V, Bondurand N, Kuhlbrodt K, Goerich firmed cause of infertility. J Med Genet 48:
Omura T: A common trans-acting factor, DE, Préhu MO, et al: SOX10 mutations in pa- 710–712 (2011).
Ad4-binding protein, to the promoters of ste- tients with Waardenburg-Hirschsprung dis- Vetro A, Dehghani MR, Kraoua L, Giorda R, Beri S,
roidogenic P-450s. J Biol Chem 267: 17913– ease. Nat Genet 18:171–173 (1998). et al: Testis development in the absence of SRY:
17919 (1992). Polanco JC, Wilhelm D, Davidson TL, Knight D, chromosomal rearrangements at SOX9 and
Murphy MW, Lee JK, Rojo S, Gearhart MD, Kura- Koopman P: Sox10 gain-of-function causes SOX3. Eur J Hum Genet 23:1025–1032 (2015).
hashi K, et al: An ancient protein-DNA inter- XX sex reversal in mice: implications for hu- Viger RS, Guittot SM, Anttonen M, Wilson DB,
action underlying metazoan sex determina- man 22q-linked disorders of sex develop- Heikinheimo M: Role of the GATA family of
tion. Nat Struct Mol Biol 22:442–451 (2015). ment. Hum Mol Genet 19:506–516 (2010). transcription factors in endocrine develop-
Muscatelli F, Strom TM, Walker AP, Zanaria E, Raymond CS, Shamu CE, Shen MM, Seifert KJ, ment, function, and disease. Mol Endocrinol
Récan D, et al: Mutations in the DAX-1 gene Hirsch B, et al: Evidence for evolutionary con- 22:781–798 (2008).
give rise to both X-linked adrenal hypoplasia servation of sex-determining genes. Nature Wang D, Huang B, Zhang S, Yu X, Wu W, Wang
congenita and hypogonadotropic hypogo- 391:691–695 (1998). X: Structural basis for R-spondin recognition
nadism. Nature 372:672–676 (1994). Salisbury TB, Binder AK, Grammer JC, Nilson by LGR4/5/6 receptors. Genes Dev 27: 1339–
Nicholl RM, Grimsley L, Butler L, Palmer RW, JH: Maximal activity of the luteinizing hor- 1344 (2013).
Rees HC, et al: Trisomy 22 and intersex. Arch mone beta-subunit gene requires beta- Warr N, Bogani D, Siggers P, Brixey R, Tateossian
Dis Child Fetal Neonatal Ed 71:57–58 (1994). catenin. Mol Endocrinol 21:963–971 (2007). H, et al: Minor abnormalities of testis devel-
Nordenvall AS, Frisén L, Nordenström A, Lich- Sax L: How common is intersex? A response to opment in mice lacking the gene encoding the
tenstein P, Nordenskjöld A: Population based Anne Fausto-Sterling. J Sex Res 39: 174–178 MAPK signalling component, MAP3K1.
nationwide study of hypospadias in Sweden, (2002). PLoS One 6:e19572 (2011).
1973 to 2009: incidence and risk factors. J Schieck M, Schouten JP, Michel S, Suttner K, Warr N, Carre GA, Siggers P, Faleato JV, Brixey
Urol 191:783–789 (2014). Toncheva AA, et al: Doublesex and mab-3 re- R, et al: Gadd45γ and Map3k4 interactions
Ottolenghi C, McElreavey K: Deletions of 9p and lated transcription factor 1 (DMRT1) is a sex- regulate mouse testis determination via p38
the quest for a conserved mechanism of sex specific genetic determinant of childhood- MAPK-mediated control of Sry expression.
determination. Mol Genet Metab 71:397–404 onset asthma and is expressed in testis and Dev Cell 23:1020–1031 (2012).
(2000). macrophages. J Allergy Clin Immunol 138: Warr N, Siggers P, Carré GA, Wells S, Greenfield
Parakh TN, Hernandez JA, Grammer JC, Weck J, 421–431 (2016). A: Genetic analyses reveal functions for
Hunzicker-Dunn M, et al: Follicle-stimulat- Schrade A, Kyrönlahti A, Akinrinade O, Pihlajoki MAP2K3 and MAP2K6 in mouse testis deter-
ing hormone/cAMP regulation of aromatase M, Häkkinen M, et al: GATA4 is a key regula- mination. Biol Reprod 94:103 (2016).
gene expression requires beta-catenin. Proc tor of steroidogenesis and glycolysis in mouse Wat MJ, Shchelochkov OA, Holder AM, Breman
Natl Acad Sci USA 103:12435–12440 (2006). Leydig cells. Endocrinology 156: 1860–1872 AM, Dagli A, et al: Chromosome 8p23.1 dele-
Parker A, Cross SH, Jackson IJ, Hardisty-Hughes (2015). tions as a cause of complex congenital heart
R, Morse S, et al: The goya mouse mutant re- Seeherunvong T, Perera EM, Bao Y, Benke PJ, Be- defects and diaphragmatic hernia. Am J Med
veals distinct newly identified roles for nigno A, et al: 46,XX sex reversal with partial Genet A 149A:1661–1677 (2009).
MAP3K1 in the development and survival of duplication of chromosome arm 22q. Am J White PC, New MI, Dupont B: HLA-linked con-
cochlear sensory hair cells. Dis Model Mech Med Genet A 127A:149–151 (2004). genital adrenal hyperplasia results from a de-
8:1555–1568 (2015). Sekido R, Lovell-Badge R: Sex determination in- fective gene encoding a cytochrome P-450
Parma P, Radi O, Vidal V, Chaboissier MC, Del- volves synergistic action of SRY and SF1 on a specific for steroid 21-hydroxylation. Proc
lambra E, et al: R-spondin1 is essential in sex specific Sox9 enhancer. Nature 453: 930–934 Natl Acad Sci USA 81:7505–7509 (1984).
determination, skin differentiation and ma- (2008). Yujiri T, Sather S, Fanger GR, Johnson GL: Role of
lignancy. Nat Genet 38:1304–1309 (2006). Suddason T, Gallagher E: Genetic insights into MEKK1 in cell survival and activation of JNK
Pearlman A, Loke J, Le Caignec C, White S, Chin Map3k-dependent proliferative expansion of and ERK pathways defined by targeted gene
L, et al: Mutations in MAP3K1 cause 46,XY T cells. Cell Cycle 15:1956–1960 (2016). disruption. Science 282:1911–1914 (1998).
disorders of sex development and implicate a Sutton E, Hughes J, White S, Sekido R, Tan J, et Zangen D, Kaufman Y, Banne E, Weinberg-Shuk-
common signal transduction pathway in hu- al: Identification of SOX3 as an XX male sex ron A, Abulibdeh A, et al: Testicular differen-
man testis determination. Am J Hum Genet reversal gene in mice and humans. J Clin In- tiation factor SF-1 is required for human
87:898–904 (2010). vest 121:328–341 (2011). spleen development. J Clin Invest 124: 2071–
Philibert P, Biason-Lauber A, Rouzier R, Pien- Tan ZP, Huang C, Xu ZB, Yang JF, Yang YF: Nov- 2075 (2014).
kowski C, Paris F, et al: Identification and el ZFPM2/FOG2 variants in patients with Zaytouni T, Efimenko EE, Tevosian SG: GATA
functional analysis of a new WNT4 gene mu- double outlet right ventricle. Clin Genet 82: transcription factors in the developing repro-
tation among 28 adolescent girls with primary 466–471 (2012). ductive system. Adv Genet 76:93–134 (2011).

Mechanism of Sex Determination in Sex Dev 2016;10:313–325 325


Humans DOI: 10.1159/000452637

You might also like