You are on page 1of 49

REVIEW

Testis Development
Juho-Antti Mäkelä,1 Jaakko J. Koskenniemi,1,2 Helena E. Virtanen,1 and Jorma Toppari1,2

1
Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku,

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


20520 Turku, Finland; and 2Department of Pediatrics, Turku University Hospital, 20520 Turku, Finland
ORCiD numbers: 0000-0001-8489-8724 (J.-A. Mäkelä); 0000-0003-2228-334X (J. Toppari).

ABSTRACT Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular
somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell
specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic
destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of
sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ
cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full
masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell
differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is
necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex
differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades,
and these are described along with the description of molecular regulation of testis development. (Endocrine Reviews 40: 857 – 905, 2019)

M ale reproductive health is largely determined


already in early development of the testis.
Differentiation of somatic cells to Sertoli cells in the
Sertoli cell identity is decisive for the future reproductive
health, and the number of Sertoli cells determines
sperm production capacity. In this review we describe
early bipotential gonad starts the male-specific devel- germ cell specification, differentiation of testicular so-
opment and guides the germ cells to the spermatogenic matic cells, and disorders that arise from defects in testis
lineage. Sertoli cells coordinate differentiation of other development.
somatic cells in the developing testis, including Leydig For this narrative review we have followed the
cells that produce testosterone required to masculinize literature with the search terms primordial germ cell,
the fetus. Normal differentiation and maintenance of testis and stem cell, or spermatogenesis.

Primordial Germ Cell Specification of specific RNAs and proteins to a certain area within
the oocyte cytoplasm, called the germ plasm. When
Specification of the germline precursors during early embryogenesis starts, the cells that inherit the germ
embryonic development is essential for perpetuation of plasm are recruited into the germline. In the epigenesis
the species, as these cells will eventually give rise to mode no maternally deposited germ plasm has been
mature gametes, sperm, and eggs, the vectors of observed, and the germ lineage is specified from a rather ISSN Print: 0163-769X
transgenerational genetic information. There are two homogeneous population of embryonic cells by in- ISSN Online: 1945-7189
distinct modes of germline specification in metazoans: ductive signals from embryonic and extraembryonic Printed: in USA
Copyright © 2019
preformation and epigenesis (). The former refers to sources. In mice, humans, and probably most other
Endocrine Society
inheritance of maternal determinants, whereas in the mammals, the germline is determined by the epigenesis Received: 14 May 2018
latter mode germ cells are specified by inductive signals. mode, and the first cells of the germ lineage are called Accepted: 17 July 2018
Preformation is based on synthesis, transportation, and primordial germ cells (PGCs). PGCs are specified before First Published Online:
consequential unequal distribution and accumulation gastrulation during early embryonic development both 26 December 2019

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 857


REVIEW

ESSENTIAL POINTS
· Cells of the germ lineage are specified at an extraembryonic location and migrate to the presumptive gonad during early
fetal development
· Induction of sex-determining region Y (SRY) expression in the bipotential gonad defines the onset of testicular
differentiation and determines the sex of the developing individual
· Specification of somatic gonadal cells to Sertoli cells determines testicular differentiation, including germ cell
development to spermatogenic cells
·· Sertoli cell identity requires constant maintenance
Defects in the genetic machinery regulating testis development cause disorders of sex differentiation
· Testicular hormones regulate development of reproductive tract and external genitalia

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


in humans (likely during the second week after fertil- of SMAD, leads to a severe reduction in the total
ization) and mice [embryonic day (E)  to ] (). In the mPGC number or their complete absence, but gene
XY embryo, they form the first distinctive sub- dosage also plays a role (–).
population of embryonic cells that can be considered Germ cell competence is initially induced by
the predecessors of spermatogonial stem cells (SSCs)— BMP in as few as six cells of the proximal epiblast on
the cells responsible for lifelong sperm production in E., just preceding the onset of gastrulation ().
men and perpetuation of the species. These founder cells are in direct contact with the
Research on early PGCs or their immediate pre- overlying ExE and they differ from their somatic
cursors has been hampered by their low number and neighbors by the expression of B lymphocyte–
embedded location among the somatic cells of the induced maturation protein- [BLIMP, officially
perigastrulation embryo. However, the mechanisms PR/SET domain (PRDM)] (, ). The ability to
and the molecular cues responsible for specification of activate BLIMP expression is not restricted to a
mouse and human PGCs have been elucidated during specific subset of epiblast cells, but it is the local high
recent years. Adoption of PGC fate embarks these cells BMP that specifies the mPGC precursors in a
on a developmental trajectory that clearly separates WNT-dependent manner (, , ). Additionally,
them from the surrounding somatic cells, and their anterior visceral endoderm secretes BMP signaling
early differentiation is characterized by three different antagonists [including cerberus  (CER) and dick-
but interconnected events: repression of the somatic kopf  (DKK)] to inhibit mPGC specification of
mesodermal program, reacquisition of pluripotent anterior epiblast cells (Fig. a) (, ). It is, however,
gene expression, and global reprogramming of the a very strictly temporally restricted process (E.~E.),
epigenome. The first two are discussed below, and the and competence of the epiblast cells to give rise
last one is discussed later in this article. In this section to mPGCs is markedly reduced after E. ().
we describe the current understanding about PGC Additionally, mPGC precursors need to express
specification in mice and humans. BLIMP at a sufficiently high level to commit to the
germ lineage ().
PGC specification in the mouse Wnt is expressed by epiblast cells and in the
posterior visceral endoderm (Fig. a). Its expression is
Signaling pathways upstream of mouse directly or indirectly activated by BMP, which has a
PGC specification crucial role (i) in priming epiblast cells to respond to
In mice, PGCs are induced from the posterior WNT signaling activation in a proper manner, and (ii)
proximal epiblast cells of the egg cylinder at ~E.. in the induction of Blimp and PR/SET domain 
Extraembryonic ectoderm (ExE)–derived bone mor- (Prdm), another mPGC specification gene ().
phogenetic protein (BMP) and BMPb, as well as BMP/SMAD and WNT/b-catenin signaling path-
BMP originating from the posterior visceral endo- ways thus work in parallel to ensure sustained ex-
derm, act on a subset of epiblast cells (Fig. a), and pression of BLIMP and its downstream targets, and
subsequently a founder population of  to  mouse PRDM, whose expression is greatly enhanced by T
PGCs (mPGCs) is observed at the base of the de- (Brachyury, officially TBXT), an effector and target of
veloping allantois in extraembryonic mesoderm of WNT signaling (Fig. b) (). Importantly, mPGC
an E. embryo (–). BMPs act via SMAD sig- formation fails in Wnt and catenin b (Ctnnb)
nal transducers, and phosphorylation of SMAD, mutants (Ctnnb encodes the WNT signaling trans-
SMAD, and SMAD triggers complex formation ducer b-catenin) regardless of BMP signaling (, ).
with SMAD and its nuclear translocation. Loss of any These data indicate that BMPs alone cannot induce or
of the involved BMPs, SMADs ,  and , but not that maintain mPGC specification to a sufficient extent but

858 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Figure 1. (a) PGC specification in the mouse. Mouse PGC precursors are induced in the E6.5 embryo from the posterior proximal
epiblast cells of the egg cylinder. BMP4 (green) and BMP8b (light green) from ExE, and BMP2 (khaki) from the posterior visceral
endoderm (PVE), plus WNT3 (orange) from the epiblast and PVE act on a subset of epiblast cells, which are recruited into the germline
as PGC precursors. Anterior visceral endoderm (AVE) secretes BMP signaling antagonists to inhibit ectopic mPGC specification in the
anterior epiblast. (b) Pathways involved in formation of mPGC precursors. ExE-derived BMP4 and BMP2 from the posterior visceral
endoderm (PVE) act via SMAD signal transducers to inhibit the expression of somatic genes and induce Blimp1 and Prdm14 in mPGC
precursors. WNT3 is directly or indirectly activated by BMP4. The concerted action of BMP/SMAD and WNT3/b-catenin signaling
pathways is needed to maintain the high expression of Blimp1 and Prdm14, and specification of mPGCs. (c) PGC specification in
humans. The epiblast of an E12 human embryo takes the shape of a disc and is surrounded by the amnion on the dorsal side and
hypoblast on the ventral side. The exact origin of hPGC precursors is not known, but they likely arise in either the posterior epiblast or
dorsal amnion in response to signals (BMPs, WNTs) derived from the epiblast, amnion, or hypoblast. Inhibitory signals are probably also
at play to prevent ectopic hPGC formation. (d) Synthesis of putative interactions involved in hPGC(LC) specification. WNT signaling
activates the expression of EOMES in hPGCLC precursors. EOMES then induces the expression of SOX17, which is also a target of BMP
signaling. TFAP2C and BLIMP1 are consequently upregulated, resulting in adoption of germ cell fate and repression of the somatic
program.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024

depend on WNT/b-catenin/T signaling for suste- (–). BLIMP-positive cells of an E. embryo
nance of BLIMP and PRDM expression. are a subset of cells that are known to express
interferon-induced transmembrane protein 
BLIMP1 as the master regulator of early mPGCs (IFITM, also known as mil- or fragilis) at E. ().
BLIMP is a DNA-binding protein that is known There is a progressive increase in their number, and at
mainly to repress but also to activate gene expression the midstreak stage (E.) ~ BLIMP-expressing

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 859


REVIEW

cells are observed in a tight cluster. At the early bud gonads devoid of germ cells and consequent sterility in
stage (E.) their number has risen to ~ cells. Tfapc-null mice of both sexes (). In vitro data show
Genetic lineage tracing experiments have revealed that Tfapc-deficient mPGCs fail to repress the so-
that all BLIMP-positive cells of the E. embryo matic program and do not upregulate germ cell
are committed to the germ lineage and are thus the markers Nanos and deleted in azoospermia-like
earliest identified definitive precursors of adult (Dazl), a situation reminiscent of and partially phe-
mouse germline stem cells (). Blimp mutant cells nocopying loss of Blimp (, ).
fail to specify as mPGCs, highlighting the impor- BLIMP, PRDM, and TFAPC collectively
tance of BLIMP for germline development (, ). constitute a transcription regulatory network essential
BLIMP not only initiates mPGC specification but for the adoption of germ cell identity in the mouse (,
it also works as a major coordinator responsible for , ). BLIMP-dependent induction of Tfapc,
adoption of mPGC-specific gene expression via direct maintenance of it by PRDM, and the mutually
repression of the somatic program and promoting interdependent expression of PRDM and BLIMP
induction of other mPGC specification genes, in- create a functional triad that is needed to ensure

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


cluding Prdm, transcription factor (TF) AP-g proper and timely specification and commitment of
(Tfapc), DND miRNA-mediated repression inhibitor mPGCs (, , ). PRDM and BLIMP are likely
 (Dnd), Kit, and Nanos (, ). The first two of the master regulators of mPGC development, whereas
these (PRDM and TFAPC) form a tripartite TFAPC appears to have an auxiliary role in mPGC
mutually interdependent transcriptional complex with formation and maintenance, and it probably modu-
BLIMP, whereas the remaining three (DND, KIT, lates the response of a subset of BLIMP and PRDM
and NANOS) are critical for the early embryonic targets (, ).
germ cell development and survival (–). Besides
BLIMP, these cells also express high levels of tissue- mPGC-like cell induction in vitro
nonspecific alkaline phosphatase (TNAP, officially mPGC-like cells (mPGCLCs) with germline compe-
ALPL), IFITM (fragilis), and developmental tence and spermatogenic differentiation ability can be
pluripotency-associated protein (DPPA) (also known derived in vitro from pluripotent stem cells (PSCs)
as stella and PGC), and—in stark contrast to their either via inducible expression of TFs or by treating the
mesodermal neighbors—display repression of Hox cultured cells with cytokines (, , ). Notably,
(Homeobox) genes, especially Hoxa and Hoxb, two there prevails a narrow window of action for mPGC-
key genes for mesodermal commitment (, –). inducing signals in vitro, too, and the transient pre-
BLIMP also mediates repression of the incipient gastrulating epiblast-like cell (EpiLC) population that
somatic program in mPGC precursors and promotes arises from pluripotent cells rather quickly loses its
acquisition of germ cell characteristics, including competence to give rise to mPGCLCs. Interestingly,
reactivation of pluripotency-associated gene expres- when the mPGCLC state is induced via introduction
sion (, , ). One mode of BLIMP action is of TFs, the nascent mPGCLCs do not display a
modification of histone methylation in a complex with transient upregulation of mesodermal genes (Hoxa,
protein arginine methyltransferase  (PRMT), and Hoxb, and T), as is the case in vivo and when the
the BLIMP/PRMT complex has been implicated in mPGC-like state is induced with cytokines (, , ,
maintenance of mPGC lineage during migration (). ). These data suggest that upon specification nascent
Blimp-deficient mPGC-like cells fail to repress the mPGCs not only activate an mPGC program but also a
somatic program (including Hox cluster activity, somatic mesodermal program, and that the former
epithelial–mesenchymal transition-associated genes, subsequently extinguishes the latter.
cell cycle progression genes, and DNA methylation Similar to their precursor cells, EpiLCs, mPGCLCs
machinery), and they inconsistently activate the have a limited lifespan, and they cannot be maintained
mPGC transcriptional signature (e.g., Dppa, Nanos) for more than  days in vitro (). Culture conditions
(). that would enable long-term maintenance of these
Subsequent to BLIMP, Prdm is induced in the cells are yet to be defined, although the requirements
mPGC precursors at E. in a BMP-dependent for their robust expansion have been recently char-
manner, and its expression is maintained by acterized (). Comparative analyses have shown that
BLIMP (, , , ). In Prdm-null mice, mPGCs mPGCLCs derived from embryonic stem cells (ESCs)
are formed at lower numbers and there is a nearly are very similar to migratory (until ~E.) mPGCs
complete loss of mPGCs by E. (). BLIMP di- (). In vitro mPGC specification is therefore
rectly induces Tfapc, which is expressed in mPGCs considered a reliable reconstitution of the in vivo
from E. onward. TFAPC exerts its effect via events and it is considered to provide a relevant model
BLIMP and is indispensable for germline develop- to analyze the mechanisms responsible for formation,
ment and maintenance (, ). Tfapc-deficient development, and maintenance of mPGCs. Among
mPGC-like cells do not migrate and are lost (prob- other things, this has enabled researchers to look into
ably via somatic differentiation) at ~E., resulting in the reprogramming of mPGC epigenome (discussed

860 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

later in this review) that would not be feasible in vivo (). In summary, these data show that BMP/SMAD
(). However, there are some obvious differences and NANOG work independently and synergistically
between mPGCLCs and in vivo mPGCs. For instance, during mPGCLC formation. These data suggest that
day  mPGCLCs express at best only low levels of there might be a number of interacting and at least
genes—such as Dazl and DEAD-box helicase  partially redundant mechanisms and pathways in
(Ddx)—that are upregulated in mPGCs upon arrival function to ensure germline commitment of a sufficient
in the primordial gonad and considered to play a role subset of epiblast cells.
in silencing of retrotransposons in the male and in
onset of meiosis in the female (–). Their low PGC specification in humans
expression in mPGCLCs is attributed to accumula- That the germline is established in humans between E
tion of repressive and lack of active histone marks at and E makes research on human PGC (hPGC)
these genes, and it may also indicate that mPGCLCs specification impracticable. Hence, knowledge con-
are more equivalent to migratory mPGCs (). A recent cerning the molecular events in pregastrulating hu-
report by Hill et al. () suggests that proper activation man embryo, where hPGCs are specified, is lacking.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


of these genes depends on a concerted action of The disc-shaped epiblast of a human post-
multiple factors, which provides an explanation for implantation embryo is sandwiched by the amnion on
why it has been difficult to recapitulate it in vitro. the dorsal side and cells of the yolk sac–contributing
hypoblast on the ventral side. This structural orga-
Reactivation of pluripotent gene expression in nization separates the putative hPGC precursors in the
the germline epiblast from trophectoderm, the functional coun-
An interesting feature in mPGC development is the terpart of the mouse ExE. It is therefore likely that the
reactivation of pluripotent gene expression. NANOG, inducing agent originates from an alternative source,
octamer-binding TF  (OCT, officially POUF), and such as the hypoblast, or is secreted by the epiblast cells
sex-determining region Y (SRY) box (SOX) form a themselves, as is the case in the rabbit, another
core gene regulatory network (GRN) and promote the mammalian species with disc-shaped epiblasts ()
pluripotent state in the inner cell mass of a blastocyst. (Fig. c). An intriguing new insight into the discussion
NANOG is re-expressed in the posterior epiblast at was introduced by Sasaki et al. (), who studied the
E., whereas SOX is detected in the anterior epiblast, specification of PGCs in the cynomolgus monkey. “Strikingly, SOX17, a critical TF
and OCT is ubiquitously expressed in the prestreak, They demonstrated that cynomolgus monkey PGCs for endoderm lineages, is
early streak, and midstreak embryos (, ). In Sox- are induced before gastrulation in the dorsal amnion essential for hPGC(LC)
deficient embryos, mPGC specification is compro- in response to signals derived from the amnion itself specification.”
mised, and SOX also plays a role in promoting and the extraembryonic mesenchyme. Given the
mPGC proliferation (). NANOG is strictly not similarity of early embryonic development between
needed for mPGC specification, but proliferation and humans and the cynomolgus monkey, these data
survival of migrating mPGCs and germline develop- suggest that hPGCs may arise mainly in the nascent
ment depend on NANOG to a certain extent (–). amnion instead of the posterior epiblast. Thus, it
In vitro, BMP induces mPGCLCs in EpiLCs in lower seems that PGCs of different mammalian species arise
numbers in Nanog mutant cells, suggesting that in distinct parts of the perigastrulation embryo in a
NANOG is required for robust mPGC(LC) formation manner that reflects diverse strategies for embryonic
(). Interestingly, recent research has demonstrated patterning in a particular species.
that a single NANOG target gene, estrogen-related Although the exact embryonic origin of hPGCs
receptor b (Esrrb), can substitute for Nanog loss in awaits clarification, subsequent human germline de-
germline development (). Surprisingly, however, velopment is rather well characterized. As reviewed by
overexpression of NANOG in EpiLCs is able to induce De Felici (), microscopic studies by Politzer (,
mPGCLC independently of BMP and WNT signaling. , ) and Witschi () identified a group of
Moreover, SOX represses mPGCLC state induction  to  large, spherical cells in the endoderm of the
by NANOG in EpiLCs, whereas it has no effect on yolk sac in a -week-old human embryo (). During
BMP-mediated mPGCLC formation (). the following days, a substantial increase in their
These data provide an important example of number was also recorded, and the size of the pre-
context-dependent action of pluripotency-associated migratory hPGC population rose to ~ cells.
factors during development: while working in an in- Markers of early [ to  weeks post coitum (wpc)]
terdependent and synergistic fashion to form the self- hPGCs include TNAP, NANOG, OCT, stage-specific
sustaining ESC core GRN, SOX is a NANOG embryonic antigen  (SSEA), and KIT (–). The
antagonist during mPGC(LC) induction. The sequence expression of DAZL, DDX, and fucosyltransferase 
and dynamics of Blimp, Prdm, and Tfapc activation (FUT) are subsequently activated or enhanced in
differ somewhat whether mPGCLC formation is in- maturing hPGCs/gonocytes ( to  wpc) (, ).
duced using BMP or overexpression of NANOG, but Notably (and in contrast to mPGCs), hPGCs do not
the derived cells are transcriptionally almost identical express SOX (). Morphologically, early hPGCs do

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 861


REVIEW

not differ much from neighboring somatic cells, but as precursors is contradictory. Recently it was shown that
they mature, they grow in size and become round or TFAPC is induced in nascent hPGCLCs by BMP
oval and have a clear cytoplasm and a large nucleus signaling, that is, independent of SOX, and it was
with one or two prominent nucleoli (, , ). postulated that TFAPC not only drives a distinctive
However, maturing hPGCs are not a morphologically developmental program but is also needed for sus-
homogeneous population of cells (). tained expression of SOX, BLIMP, NANOG, OCT,
Although experimental and definitive data are and NANOS (). This is in stark contrast to its
lacking, it has been proposed that a subset of posterior somewhat minor role during mPGC specification ().
epiblast cells (or cells of the dorsal amnion) are Other reports imply that SOX and BLIMP act
specified as hPGC precursors between E and E in upstream of TFAPC for hPGCLC specification (,
response to autocrine and/or paracrine signals (BMP ). Both SOX and BLIMP were shown to be
and WNT) (Fig. c). During the following days these necessary for hPGCLC formation, but SOX alone
cells move out of the embryo proper and are tran- was able to induce germ cell genes independent of
siently incorporated into the extraembryonic meso- cytokines in i-cultured germ cell–competent hESCs

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


derm. At E they are found near the base of allantois (). A novel regulatory network composed of OCT,
in the wall of the definitive yolk sac, and upon ini- paired box  (PAX), and BLIMP was also recently
tiation of TNAP expression are considered true implicated in hPGC induction (). How all of these
hPGCs (). TFs interact and to what extent they display re-
dundancy is not well understood at present. BMP
Induction of human PGCLCs and BMP (but not BMP or BMPa) work in-
The basic mechanisms of hPGC specification have terchangeably in induction of hPGCLCs, whereas SCF,
been elucidated in vitro. Although EpiLCs are readily epidermal growth factor, and LIF have an auxiliary role
induced from murine ESCs by Activin A and basic in the process and they support the maintenance of
fibroblast growth factor (bFGF), the propensity of nascent hPGCLCs () (Fig. d).
human ESCs (hESCs) for human PGCLC (hPGCLC) When hiPSCs are used as a source of hPGCLCs, the
formation in the presence of bFGF is poor, and the cultured cells first give rise to incipient mesoderm/
approach used for mouse cells fails to render hu- primitive streak-like cells (iMeLCs), which modestly
man PSCs competent for the germline (, –). and transiently upregulate genes characteristic of me-
Nonetheless, hPGCLCs can be induced from hESCs sodermal lineages [including T and eomesodermin
and human-induced PSC (hiPSCs), and robust culture (EOMES)] (). In the presence of BMP, iMeLCs then
systems for the derivation of hPGCLCs have been robustly generate hPGCLCs. Recently it was shown that
available since  (–). Either hESCs cultured the germline competence in iMeLCs depends on ap-
in a medium containing inhibitors against glycogen propriate dosage and duration of WNT signaling (, ).
synthase kinase b (GSKB), c-Jun N-terminal kinase WNT signaling activates EOMES, which in turn induces
(JNK), MAPK kinase (MEK), and p (5 i culture) the hPGCLC specification program by upregulating the
or hiPSCs stimulated by Activin A and a WNT sig- expression of key genes, most notably SOX.
naling agonist (ACTA plus CHIR) display high At the moment, there is no consensus as to what is
competency for germline fate and, in the presence of the hierarchy of different TFs during hPGC(LC)
BMPs, stem cell factor (SCF), epidermal growth factor, specification, which TFs are absolutely required for
and leukemia inhibitory factor (LIF), they robustly give the process, and what is the relative importance of the
rise to hPGCLCs (, ). The transcriptional profiles TFs involved. There are at least two plausible expla-
of hPGCLCs are highly similar independent of the nations for the discrepancies in the available data: first,
induction method or the starting material (hESCs or the cells of origin (i-cultured hESCs vs hiPSCs cul-
hiPSCs) (). tured under a conventional condition) probably bear
A shared feature in PGC(LC) specification between distinct states of primed pluripotency that might affect
humans and mice seems to be a dependency on a their route to germ cell fate; and second, both ablation
signaling network that is orchestrated by three key and excessive overexpression of TFs might blur the
proteins. However, as described in more detail below, physiological significance of the findings. Future
the identity of the proteins involved and, for the studies that precisely define the function of key TFs
proteins that are shared, their significance for robust in a physiologically meaningful way are warranted.
PGC specification depends on the species. Strikingly,
SOX, a critical TF for endoderm lineages, is essential PGC specification in humans vs mice
for hPGC(LC) specification (, , ). It is the Many aspects of mammalian PGC specification seem
earliest known marker for hPGCLCs and works up- to have been conserved in evolution, but there are also
stream of BLIMP () (Fig. d). Sox is also induced obvious differences in the mechanisms responsible for
in the mouse when the germ lineage is established, but PGC specification in humans and mice. What then
it is dispensable for mPGC specification (, ). Data might explain the observed dissimilarities? First, the
concerning the induction of TFAPC in hPGCLC early postimplantation development of mouse and

862 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

human embryos at the time when PGCs are formed In summary, in vitro–specified PGCs faithfully
may explain such differences: the human embryo is a mimic most of the key events that are associated with
bilaminar flat disc, whereas the mouse embryo forms a PGC formation in vivo: repression of somatic genes,
cup-shaped egg cylinder (). Second, in the early reactivation of pluripotency gene expression, and global
postimplanation embryo of the mouse, the PGC- reprogramming of the epigenome. Owing to epigenetic
inducing ExE is situated on top of the epiblast, but regulation, implementation of the germ cell–specific
an equivalent structure does not exist in humans (). transcriptional program is incomplete (as in early PGCs
Third, mouse and human PSCs, which are the in vitro in vivo), which might reflect lack of an appropriate
source of PGCLCs, differ in their regulation, growth environment, that is, similar to the one that PGCs are
requirements, and cell morphology (). Human PSCs exposed to upon and after integration into the gonadal
depend on bFGF for long-term maintenance, whereas primordium (, , , ). It is also possible that
mouse PSCs rely on LIF to support the pluripotent hPGCLCs correspond to early postspecification hPGCs,
state. Human ESCs actually resemble more (in terms and similar to mPGCLCs do not express germ cell–
of cytokine dependence, epigenetics, and tran- specific genes DDX and DAZL, and display just some

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


scriptomics) mouse epiblast stem cells (EpiSCs), an but not all aspects of epigenetic reprogramming (, ).
epiblast cell-like cell line that is refractory to PGC Further maturation of hPGCLCs probably depends on
induction, than mouse ESCs (, –). identifying the proper cytokines that enable hPGCLCs
Human PGCLCs are transcriptionally close to to proceed on their developmental pathway.
hPGCs of -week-old male human embryos (corre-
sponds to E. to E. in the mouse) but, similar to PGC homing
mPGCLCs, express a significantly lower level of germ As described in the previous section, PGC precursors
cell and meiosis-related markers, such as DDX and are likely induced in the human embryo within the
DAZL (, , ). In addition to the critical role of epiblast or dorsal amnion during the second embryonic
SOX, the expression level of SOX is another ob- week. In human, PGCs are initially observed at an
vious difference between human and mouse PGC(LC)s. extraembryonic location on E and the first PGCs
SOX is rapidly downregulated in human PSCs colonize the gonadal ridges (GRs) during the fifth
destined to germ cell fate and it is undetectable in week postconception (, ). Hence, PGCs need to
“…the fate of mismigrated
hPGCLCs and hPGCs but has an important role in translocate a considerable distance to contribute to the
PGCs depends not only
mPGC specification and proliferation (, , ). In- developing bipotential gonad (Fig. a). In mammalians on prosurvival factors but
terestingly, BLIMP, working downstream of SOX, they reach gonads by both active and passive means, also on environmental
may not be as important in humans as in mice (). that is, migration and displacement caused by em- permissiveness.”
Moreover, the role of PRDM in hPGC(LC) speci- bryonic growth movements (, ). A four-step ap-
fication is unclear, as PRDM expression is very low in proach can be used to describe PGC homing: (i)
nascent hPGCLCs and it is only weakly induced during transepithelial, (ii) dorsal, and (iii) lateral migration
the following few days (, ). PRDM is repressed in followed by (iv) coalescence with somatic cells to form
late hPGCs, and its overexpression is associated with the complete embryonic gonads (Fig. b). To what
delayed differentiation of hPGC(LC)s and germ cell extent the displacement of PGCs is due to active
tumor (GCT) formation (). The interdependent (migration) and passive (folding, growth, and devel-
BLIMP-PRDM-TFAPC GRN, which is needed for opment of embryonic structures) mechanisms is a
robust specification of mPGCs, thus does not seem to matter of debate, and the whole concept of germ cell
exist in human. Moreover, Krüppel-like factor  migration during embryonic development has been
(KLF), a gene associated with naive pluripotency, questioned (). However, many lines of evidence
is sharply upregulated upon hPGCLC, but not suggest that PGCs are migratory cells: PGCs are motile
mPGC(LC), specification (, , , ). cells both in vivo and in vitro, mouse GR explants attract
Available pieces of data suggest that hPGCs are isolated early PGCs over long distances in vitro, and
induced from a transient peri-gastrulating mesoderm- ectopic expression of a putative PGC attractant results
like state (, ). The molecular bases of human and in aberrant germ cell migration in vivo (–). The
mouse PGCLC induction and in vitro maintenance are conclusion from these studies and numerous other
similar, but the transcriptional programs downstream reports is that PGCs actively migrate in the developing
of induction display considerable divergence be- embryo, at least over relatively short distances ().
tween the two species and depend partially on dif-
ferent signaling cascades. Most strikingly, a clearly Route of PGC gonad-oriented displacement
noticeable transient induction and subsequent re- At the time of PGC specification, the prospective
pression of the somatic mesodermal program is gonads near the developing root of the mesentery exist
observed only during mPGC(LC) induction, and it is as slightly condensed structures of the coelomic epi-
probably needed for proper activation of the tri- thelium (CE). To build a functional gonad, PGCs need
partite transcriptional network that does not exist in to be translocated from the yolk sac endoderm to
humans (, ). the embryo proper, and the somatic precursors of

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 863


REVIEW

Figure 2. (a) Site of nascent hPGCs. hPGCs are observed near the base of the developing allantois in the wall of the definitive yolk sac
that is outside the embryo proper, soon after their specification. As the embryo folds, the part of extraembryonic endoderm that is
inhabited by the nascent PGCs is incorporated into the embryo and forms the midgut and the hindgut. (b) PGC displacement. The
route of PGC migration/displacement during early fetal development is shown. A four-step approach can be used to describe PGC
homing: (i) transepithelial in the endodermal epithelium, (ii) dorsal through the mesenchyme of the gut mesentery, and (iii) lateral in
the mesoderm, and finally (iv) coalescence with somatic cells of the GRs to contribute to the embryonic gonads.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024

864 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

the gonad need to proliferate and reorganize to form the notion of nerve fibers playing an important role
the GRs, the primordia of the future testis or ovary. in directional guidance and promotion of PGC
Roughly  week after specification at an extraem- survival (, , ). Research done in mice suggests
bryonic location, PGCs get reincorporated into the that PGC survival can only be supported within the
embryo proper as the embryonic disc folds and the migratory path, and if PGCs fall outside this tra-
portion of the yolk sac that hosts PGCs becomes jectory, cell survival–promoting signals are too weak
the hindgut and the midgut. Consequently, PGCs can to maintain these ectopic PGCs and they undergo
be found as single cells near the aorta among the apoptosis (, , ). Interestingly, not all stray
endodermal cells of the primitive hindgut and PGCs perish, and mPGCs that end up in the adrenals
midgut epithelium. These events take place during produce oocytes and enter meiosis independent of
the fourth embryonic week and precede the for- the sex (). It has also been speculated that PGCs
mation of GRs. might contribute to the pool of adult-type hema-
If indeed PGCs were to actively migrate from the topoietic stem cells that emerge in the aorta–
yolk sac to the GR, they would first need to push gonad–mesonephros region during embryogenesis

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


endodermal epithelial cells out of their way within the (). Thus the fate of mis-migrated PGCs depends
yolk sac (stalk) and in the primitive gut (i), then not only on prosurvival factors but also on envi-
penetrate into to surrounding mesenchyme and move ronmental permissiveness. Owing to a distinctively
dorsally through the gut mesentery (ii), and, upon different epigenetic status, it is unlikely that the stray
arriving at the preaortic region in the midline of the PGCs could contribute to somatic tissues, with he-
abdominal dorsal body wall, move a little farther in the matopoietic stem cells probably being the remarkable
lateral direction (iii) to make it to the GR (iv) (Fig. b). exception.
Recent and previous research also question the axiom In the mouse, migratory PGCs are mostly found
of long-range PGC migration and provide data to in tissues as single cells but they occasionally extend
support passive displacement of PGCs from the long processes to other PGCs and might move
endoderm to the GR (, ). However, these data within a tissue as a group of cells (). The molecular
support the notion of short-range migration of PGCs control of PGC migration is another much-debated
at particular steps of homing, for instance, when topic. To what extent is the proper guidance based on
PGCs cross the basal lamina of the endodermal attraction, and what is the role of repulsive cues? It
epithelium to enter the mesenchyme. This particular has been suggested that mediators such SCF
step might involve epithelial-to-mesenchymal and fibronectin, which are abundantly expressed
transition, a process that converts well-organized along the migratory route, and adhesion molecules
and compact epithelial cells into migratory mes- expressed by PGCs themselves are responsible for
enchymal cells. early (before the fifth week of embryonic develop-
ment) guidance of PGCs prior to establishment of the
Physical guidance of PGC migration connections of the nervous system (, , , ). If
It has been suggested that nerve fibers serve to guide this hypothesis holds true, once the nerve connec-
the movement of PGCs from the dorsal hindgut tions are established, the chance of going astray
mesentery via the preaortic region to the gonadal would be considerably diminished. Besides providing
anlage (). Indeed, both (first) hPGCs and nerve a physical lifeline for PGCs, the nervous system
fibers arrive at GRs during the fifth embryonic week might also use other means to aid PGC homing, and
(). There is, however, a wide spatial distribution of Schwann cells have been suggested to secrete
hPGCs during the th to th wpc, and during de- lysophosphatidic acid, which attracts and guides
velopment some hPGCs are still located in the gut PGCs ().
epithelium, whereas others are already enclosed within The route of PGC homing is virtually identical
the nascent testis cords (, , ). These observations in humans and mice. Migrating PGCs move in an
bring up many questions that have been insufficiently amoeboid fashion and have a dynamic cell mor-
addressed to date. What is the reason for the wide phology, broad leading edge, and low adhesiveness and
spatial distribution of hPGCs within the developing is therefore reminiscent of immune cell migration ().
embryo/fetus? What is the latest time point of gonadal Surprisingly, migratory PGCs proliferate and have
entry? What is the mechanism that drives gonad- been suggested to depend on anaerobic energy pro-
oriented movement of hPGCs at later time points? duction (). However, many of the genes that are
What is the destiny of hPGCs that fail to reach the enriched in migratory hPGCs, when compared with
gonad and what is the initial cause for improper later fetal germ cells, are associated with aerobic
displacement? respiration (). PGC proliferation and survival/
If ectopic PGCs are not cleared by apoptosis, they apoptosis are controlled by a number of genes, as
might give rise to GCTs (). In humans, extragonadal recently reviewed by Hamer and de Rooij (). The
GCTs are most commonly found along the midline molecular control of PGC migration has been char-
and within the central nervous system, which supports acterized only in the mouse.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 865


REVIEW

Molecular mechanisms of PGC migration loss of Cdh results in mis-migration and ectopic
Although the molecular mechanisms responsible for PGCs, Itgb-deficient mPGCs colonize the GRs
attracting, guiding, and, finally, stopping hPGCs are with a poor efficiency (, ). A role for ITGB in
largely unknown, there is a handful of molecular the mPGC migratory process is also supported by
players that have been shown to regulate mPGC the presence of fibronectin, an ECM molecule that
trafficking. Molecular regulation of embryonic germ interacts with ITGB, along the route of mPGC
cell migration in different species has recently been migration. Fibronectin is also required for mPGC
reviewed by Barton et al. (). Although numerous displacement in vitro (). Additionally, TGFb and
mechanisms—chemotactic movement based on tissue- FGF signaling have been implicated in mPGC mi-
wide gradient of an attractant, dynamic adhesion to gration (–).
close-by PGCs and extracellular matrix (ECM) For successful homing of PGCs, an equally im-
components along the migration path, lipid portant question as to what induces and guides PGC
modification-mediated PGC repulsion, other repulsive migration is what eventually stops it. It has been
cues, and neuronal guidance—have been suggested to presumed that PGCs become nonmotile upon co-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


be involved in PGC homing, the significance of dif- alescing with somatic gonadal cells. However, de-
ferent mechanisms in mammalian species is not well scendants of PGCs display motile properties later in
understood (, ). In mice, the data supporting life; that is, in the fetal/perinatal period as gonocytes
involvement of C-X-C motif chemokine ligand  migrate to the basal lamina of the testis cords, and in
(CXCL)/C-X-C motif chemokine receptor  the adult as SSCs constantly move within the basal
(CXCR), SCF/KIT, cadherin  (CDH), and ITGB compartment of the seminiferous epithelium
(integrin subunit b) in PGC migration is, however, (–).
relatively solid. By E. postmigratory mPGCs/early gonocytes
CXCL12/CXCR4. CXCL expressed in the become nonmotile and fail to migrate even when
body wall mesenchyme and GR and its cognate transferred into a permissive environment (). The
receptor CXCR on mPGCs form the best charac- mechanism responsible for stopping PGC migration
terized system taking part in guidance of PGCs (, once they have reached the gonad is yet to be char-
, ). Although being dispensable for the trans- acterized. It is, however, well documented that upon
epithelial migration of PGCs, it is needed for proper arriving to the GRs, the transcriptome of PGCs is
homing of PGCs in the mesenchyme. Lack of either altered: for instance, markers of an adult germline cell,
Cxcl or Cxcr results in very few intragonadal Ddx, Dazl, and germ cell nuclear acidic protein
PGCs, whereas ectopic expression of CXCL leads (Gcna), are induced, whereas Tnap, Ssea, Blimp, and
to reorientation and aberrant migration of mPGCs Kit are downregulated (–). A similar process
(, ). Despite that the CXCL/CXCR system takes place in humans, and hundreds of genes are
has been suggested to protect PGCs from cell death, differentially expressed between migrating hPGCs and
the main role for survival promotion of PGCs mitotic germ cells that have settled in the gonads (,
has been assigned to SCF/KIT signaling (, , , ). These data challenge the notion of gonocytes
, ). being equivalent to PGCs and representing just a
SCF/KIT. The SCF/KIT system has also been nonmotile version of them (). A dozen master
shown to be important for continued motility of regulators of gene expression networks, including SIX
mPGCs (). Despite being expressed along the homeobox (SIX), lysine demethylase A (KDMA),
migratory pathway both in humans and mice and and SOX, have been implicated in adoption of
forming a gradient with the highest level of ex- gonadal hPGC/early gonocyte transcriptional signa-
pression in the GR, the role of SCF as a directional ture ().
cue has been questioned (, , , ). WNTA/ If indeed a chemotactic gradient is the main
receptor tyrosine kinase–like orphan receptor  determinant of PGC gonad-oriented movement,
(ROR) signaling has been suggested to work in then reaching the highest concentration of an at-
synergy with SCF/KIT system and potentiate the tractant (i.e., the GR) would provide a natural stop
mPGC response to SCF (). Lack of either Wnta sign for PGCs. However, cessation of direc-
or Ror results in fewer mPGCs colonizing the GRs tional movement is one thing and loss of motility is
(). ROR is expressed by postmigratory human another. To contribute to a new organ, the inherent
germ cells in an -week male fetus, but it is not motile behavior of PGCs has to be repressed. How
known whether it plays a role in the migration of it mechanistically takes place is not known, but it
hPGCs (). has been suggested that cell–cell adhesion be-
Cell adhesion. Two adhesion molecules, CDH tween PGCs, as well as PGCs and gonadal somatic
and ITGB, also contribute to mPGC homing and cells, has a key role in suppression of PGC motility
are needed specifically to aid in exit from the hindgut (). The molecular events in charge of gonadal
(–). CDH is a cell–cell adhesion molecule, somatic cell specification are the topic of the fol-
whereas ITGB interacts with the ECM. Whereas lowing section.

866 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Testis Determination: Genetics involving SRY account for only % to % of all cases
and Embryology of ,XY complete gonadal dysgenesis (). As dis-
cussed later in this section, more than a dozen other
Gonads of an adult individual, testes or ovaries, have genes have been associated with ,XY gonadal dys-
their origin in GRs, two long and thin embryonic genesis (). Spontaneous mutations, deletions,
structures that run craniocaudally on the dorsal sur- duplications, or translocations in humans and in-
face of the coelomic cavity, overlying the mesonephri. troduction of transgenes into mice have shown that
Onset of CE cell proliferation in E. mice or  to  either SRY or SOX alone is sufficient to induce testis
wpc humans is the first sign of presumptive gonad development in an XX embryo (–). Among
formation. Subsequently, the underlying basal lamina the SOX family genes, SOX and SOX have also
partially disintegrates to allow migration of the mitotic been associated with human testicular or ovotes-
progeny into the dorsal inner mesenchyme to form ticular ,XX DSDs (, ).
areas of cellular condensation, later observed as the
GRs. All of the cells in GRs (supporting cells, ste-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


Formation of the bipotential gonad
roidogenic cells, germ cells) are thought to be capable Located on the medial surface of the urogenital ridge,
of differentiating into either testicular or ovarian the GR becomes morphologically distinct in human
lineages. This bipotentiality of cell fates makes GRs embryo during the fifth developmental week. GRs
unique primordia in organogenesis. arise within the intermediate mesoderm as a result of
The bipotential gonad and adrenal gland both arise CE proliferation and ingression. Proliferation of CE
from a specific region of intermediate mesoderm in the cells starts at ~E. in the mouse and these cells ul-
early embryo, the adrenogonadal primordium (AGP). timately give rise to most of the somatic cell types in
The primordial adrenal gland can be seen as a distinct the presumptive testis or ovary (, ). CE can thus
structure at ~ days postconception (dpc) in humans. be regarded as a source of multipotent progenitor cells
In the ,XY embryo the developing gonad remains (). Mitotic activity of CE cells and their ingression
bipotential until  dpc when the expression of Y into the interior of the tissue result in formation of
chromosome gene SRY is transiently induced in a thickened, elongated, and thin structures, the GRs, by
subset of somatic cells of the gonadal primordium E. in the mouse—the time of Sry activation and
(). This process, called sex determination, precedes arrival of PGCs (Fig. a). Cells of the GR contribute to “…SRY is merely a trigger and
by a couple of weeks the onset of sex differentiation the future gonads, but cell migration from elsewhere in does very little per se to
that depends on induction of steroidogenic gene ex- the embryo—at least vascular endothelial cells (VECs) promote entry into the male
pression and production of testosterone in the de- from the neighboring mesonephros, macrophages pathway….”
veloping male. Development of external genitalia and from the yolk sac or fetal liver, and germ cells from the
phallic growth are among the earliest signs of an- base of allantois—is also needed for complete testicular
drogen action. Testosterone synthesized by fetal organogenesis.
Leydig cells is also needed to stabilize Wolffian
structures, whereas Sertoli cell–derived anti-Müllerian TFs essential for GR formation
hormone (AMH) acts to regress the Müllerian ducts, Wilms tumor  (Wt) is among the earliest genes to be
the source of the fallopian tubes, uterus, and upper expressed in the mouse urogenital ridge at E ().
vagina in females (). Timelines of prenatal testis WT-positive cells of the gonadal primordium sub-
development in mice and humans are provided in Fig. sequently contribute to Sertoli cells and at least two
a and b. different interstitial cell lineages (). E. can be
The inability of a ,XY embryo to activate the considered the earliest stage of organogenesis of the
expression of SRY in a temporally, spatially, or mouse gonads or GRs. A number of genes have been
quantitatively proper manner results in a disorder of implicated in GR formation. However, the phenotype
sex development (DSD). DSDs are relatively rare of Wt, GATA-binding protein (Gata), empty spi-
congenital conditions that are characterized by atypical racles homeobox  (Emx), LIM homeobox  (Lhx),
development of chromosomal, gonadal, and ana- and nuclear receptor subfamily  group A member 
tomical sex. DSDs have their origin in complete or (Nra)–mutant mice is most severe, suggesting that
partial gonadal dysgenesis, or in undervirilization these factors are absolutely crucial for the earliest steps
or undermasculinization due to defective androgen of gonadal development (–). Nra [also
synthesis or action. ,XY complete gonadal dysgen- known as steroidogenic factor  (Sf)] expression is
esis is defined by female external genitalia, well- induced coincidentally with the onset of CE cell
developed Müllerian duct–derived structures, and proliferation in a WT/LHX-dependent manner
streak gonads. ,XY partial gonadal dysgenesis is (). Similar to the mouse, WT and NRA are
characterized by varying degrees of masculinization of expressed very early in the human presumptive gonad
the external genitalia, coexistence of Müllerian and and their expression precedes that of SRY by ~ days
Wolffian structures, and incomplete testis formation. (, ). WT is indispensable for the activation of
The etiology of ,XY DSDs is variable, and mutations Nra and Dax (officially Nrb) expression, and its

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 867


REVIEW

Figure 3. (a) Formation of the testis and timeline of mouse prenatal testis development. CE cells start to proliferate at ~E9.5 when
Nr5a1 is also induced. Many signaling pathways converge on the level of NR5A1. The underlying basement membrane disintegrates and
the cells ingress into the interior of the tissue, resulting in formation of the GRs by E10.5 when Sry is induced. The first PGCs have already
arrived in the GRs at this stage. Pre-Sertoli cells are subsequently specified and testiculogenesis starts. Sertoli cells orchestrate the
process. Other somatic populations are either recruited from the undifferentiated precursors or they migrate from elsewhere, such as
VECs from the mesonephros (MN). Sertoli cells encase PGCs into protocords. Fully developed testis cords are first observed at E13.5. (b)
Timeline of first and early second trimester testis development in humans. (c) Regulation of SRY expression in (i) the mouse and (ii)
humans. (i) In mice, a modular approach can be used to gain an insight into the complex interplay of factors involved in induction of Sry.
Modules 1 through 3 converge on a key TF that directly takes part in regulation of Sry expression. Epigenetic regulatory mechanisms
constitute module 4 that controls the access of TFs to the Sry gene and its regulatory elements and enhancers. (ii) In humans, the same
key TFs are responsible for the activation of SRY expression as in the mouse but, although many similarities can be observed,
considerable species-dependent differences likely exist.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024

loss results in an early block in gonadal development, steroidogenic tissues and die soon after birth due to
lack of reproductive organs, and late embryonic le- adrenal insufficiency (). GRs initially develop in
thality (). Nra-null mice do not develop these mice but they subsequently regress due to

868 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Figure 3. (Continued)

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


apoptosis (). These data suggest that NRA is not migrate through the basement membrane (). In the
needed for the onset of gonadal development but absence of GATA, Nra and Lhx are not induced
rather for the maintenance and differentiation of and the CE shows no features of gonadal differenti-
somatic cells within the GRs. Many signaling pathways ation (). Chromobox homolog  (CBX) also
converge on the level of NRA (Fig. a), and ac- works upstream of Nra during early gonadal de-
cordingly misregulation of Nra is associated with velopment, and Cbx-null mice display early gonadal
most mouse mutant embryos that display impaired growth defects, although normal CE proliferation
gonadal development. (, ). The expression of Wt and Emx is not
GATA expression in the CE precedes the onset of affected in Cbx-null gonads at E., but that of
epithelial proliferation (). In conditional Gata Gata, Nra, and Lhx is dramatically decreased,
knockout mice, CE cells proliferate less and integrity of suggesting that CBX exerts its effect on gonadal
the basement membrane is maintained. The cellular development via GATA and NRA.
changes driving the thickening of CE do not thus Nra expression is also regulated by SIX and
occur, and the CE of these mice remains as a mor- SIX, and Six/ double-mutant mice have small
phologically undifferentiated monolayer. This is re- gonads and complete loss of NRA-expressing cells.
lated to Emx deficiency, which results in early failure Gonadal expression of Wt, Gata, and Lhx, however,
of GR development due to inability of CE cells to is not affected, implying that loss of Six/ specifically

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 869


REVIEW

impinges on Nra (). PBX homeobox  (PBX) either permit or inhibit access to the promoter and
and insulin and insulin-like growth factor signaling regulatory elements of Sry.
have also been implicated in Nra regulation at the Next, we use a similar approach and immerse into
time of GR formation (, ). In conclusion, many the function of these modules in detail and aim at
factors participate in the timely and proper activation providing an organized description of SRY regulation
of Nra in the CE (Fig. a), and formation of (Fig. c). Despite obvious differences between humans
NRA-expressing progenitor cells represents the and mice in the structure of the SRY gene itself or in
initial stage of gonadal growth and development in the regulatory regions that surround it, as pointed out
both sexes. by Larney et al. (), owing to scarcity of research
According to current knowledge, gonadal pri- material and at the risk of irrelevant findings, we still
mordia of XY and XX embryos are indistinguishable have to resort to studies conducted with mice to
until induction of SRY at  dpc and E. in humans understand how mammalian male development is
and mice, respectively, and comprise an unorganized initially induced.
mixture of somatic and germ cells (Fig. a). Although

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


the somatic cells of embryonic gonads actively pro- Module 1
liferate in both sexes, activation of SRY expression is GATA works at the heart of module . At least three
accompanied by a proliferative burst and accelerated different factors or signaling cascades control the
growth of the testis when compared with the age- action of GATA. The most intimate of these is the
matched ovary (). zinc finger cofactor friend of GATA (FOG, officially
ZFPM), and loss of either Fog or elimination of
SRY activation and entry into the male direct interaction between the two results in reduced
developmental pathway expression of Sry, loss of Sox induction, and a con-
SRY expression is induced in human embryonic gonad sequent XY sex-reversal phenotype (). The role of
at ~ dpc and in the mouse at E. (, –). GATA action in sex determination is highlighted by
There are a few aspects of SRY that deserve special the fact that loss of Gata induced at E. (the time of
attention. First, both in humans and mice, SRY is only Sry induction), but not at later time points, causes
transiently expressed. Whereas in the mouse Sry ex- an early block in Sertoli cell differentiation and con-
pression returns to the preinduction level by E., in current male-to-female sex reversal (). GATA
the human embryo SRY expression is downregulated binding to Sry promoter does not depend solely on
at ~ dpc and it reaches the baseline transcript level interaction with FOG but also on a complex MAPK
by  dpc (, , ). Second, there is a specific (mitogen-activated protein kinase) signaling cascade
window of induction for SRY expression probably in that results in phosphorylation of GATA (, ).
both species, but experimental data show that if Sry is GATA is directly phosphorylated by p, and si-
not upregulated by E., the bipotential gonad will multaneous inactivation of pa and pb MAPKs
assume the ovarian fate (). Third, SRY is merely a leads to reduced Sry expression and gonadal sex re-
trigger and does very little per se to promote entry into versal (). GATA phosphorylation and testis de-
the male pathway and depends on its downstream velopment is also impaired in MAPK kinase kinase 
factors (most notably SOX) for initiation of the (Mapk)– and growth arrest and DNA damage-
genetic program that ultimately leads to testis de- inducible g (Gaddg)–mutant embryos (–).
termination and male sex differentiation. Fourth, it is MAPK and GADDg are thought to function up-
not just the timing but also the intensity of expression, stream of p MAPKs, and reduced phosphorylation of
and SRY abundance needs to cross a specific threshold GATA and low expression of Sry are associated with
level for proper induction of the downstream ma- reduced levels of phosphorylated p (, ). In-
chinery (). terestingly, Gaddg deficiency can be compensated for
There are at least a dozen factors that take part in by overexpression of MAPK, suggesting that these two
regulation of Sry expression in mice in a partially factors interact in the same cellular pathway during
interdependent, synergistic, or redundant fashion (Fig. gonadal sex determination. The role for GADDg in
c). Larney et al. () have proposed a modular the activation of GATA via the MAPK cascade is
approach to provide a coherent model that would supported by the fact that Gaddg expression shares
explain how these factors are functionally organized two key features of Sry expression, that is, narrow
upstream of Sry. According to the model, each module temporal window and center-to-pole spatial expansion,
is centered upon a key TF (GATA, NRA, and and, importantly, it is detected earlier than Sry ().
WT) that interacts with other proteins. The inter- There are no known cases of human XY DSD due
acting factors have complex functions and they might to GADDg or MAPK mutations, but single
act as coactivators, posttranslational modifiers, or nucleotide changes in a related protein, MAPK,
upstream regulators of primary factors. A fourth are associated with testis dysgenesis and persistence
module, which is somewhat different in nature, is of Müllerian derivatives (). Interestingly, mice
provided by epigenetic regulatory mechanisms that lacking the cognate gene display overtly normal

870 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

testis development, implying species-specific dif- derivatives of the AGP: testis, ovary, and the adrenal
ferences in involvement of MAPK family members gland (). Nra is induced very early in the mouse
in SRY regulation (). gonadal primordium (~E.), but upon sex de-
GADDg also provides a putative link to insulin/ termination its expression becomes sexually di-
IGF signaling and their role in sex determination. morphic (). In the testis, Sertoli cells and Leydig
Constitutive ablation of either via loss of insulin or the cells continue to express high levels of NRA,
IGF receptor are associated with growth and differ- whereas in the ovary it is transiently downregulated
entiation defects of the AGP, adrenal gland agenesis, and reactivation of its expression foreshadows the
and male-to-female sex reversal (, ). Although it onset of steroid hormone production ().
is not clear whether the sex differentiation defect in CBX, a core subunit of multiprotein polycomb
these mice is due to a direct (insulin/IGF signaling repressive complex  (PRC), is a chromatin modu-
being integrated into one or more of the modules) or lator that is associated with male-to-female sex reversal
an indirect (general proliferation defect with sub- in both humans and mice (, ). CBX is
sequent reduction in the number of pre-Sertoli cells) expressed in the human embryonic testis at the time of

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


mechanism, implicit data suggest that insulin/IGF SRY induction, implicating a role in early testis de-
signaling might be integrated into module  via velopment (). PRC takes part in posttranslational
GADDg (). Homeoprotein TFs SIX and SIX modification of histones and is classically associated
are also involved in Sry regulation via modules  and . with a transcriptionally repressive state of many genes,
They directly regulate the expression of Fog and but in the embryonic gonad it (or at least its CBX
Nra (the key TF of module ). SIX and SIX display component) is also implicated in activation of gene
at least partial functional redundancy, as loss of Six expression (). Research performed on a human
or Six alone does not affect embryonic gonadal Sertoli-like cell line suggests that CBX directly
development, but loss of both leads to reduced Sry regulates a number of genes involved in testis de-
expression and impaired testicular differentiation termination or associated with DSD, including NRA
(). and SOX (). Notably, SRY, GATA (as has been
The role of GATA in SRY regulation in humans is shown in the mouse), or WT do not appear on the list
unclear. Whereas GATA alone poorly activates the of CBX-upregulated genes (, ). The conclusion
SRY promoter, another well-known SRY regulator, from the existing data is that CBX exerts its effect on
“…WT1 is the most important
WT, potentiates its effect, as discussed later (). regulation of SRY via stimulation of NRA expression regulator of SRY expression in
Importantly, however, note that the complexity of (, , ). This is a common theme for other humans.”
GATA regulation (phosphorylation and interaction factors of module , too. These are SIX/SIX, LHX,
with FOG) might explain a lack of an effect in in vitro and CREB-binding protein/p-interacting trans-
promoter transactivation assays. Although GATA activator, with Glu/Asp-rich carboxyl-terminal do-
mutations have been associated with ,XY DSDs, main (CITED).
cardiac defects are the most common clinical feature of As discussed above, Nra is a direct target of SIX/
GATA mutation carriers (). It has been specu- SIX (). It is therefore likely that both CBX and
lated, however, that the mutations in these patients do SIX/SIX work in many at least partially redundant
not interfere with the ability of GATA to interact and ways to ensure sufficient activation of Sry transcription.
synergize with other proteins (WT, NRA, FOG) LHX is another sex-independent early marker of the
upon SRY regulation. GATA and FOG are expressed presumptive gonadal region of E. mouse urogenital
in the  wpc human embryonic gonad, suggesting a role ridges (). Expression of Nra is dramatically re-
in early gonad development (). duced in Lhx-null mice, and they consequently display
complete gonadal agenesis (). Interestingly, Lhx is a
Module 2 putative target of CBX in mice but not in humans (,
The second group of factors implicated in regulation ). CITED is a context-dependent transcriptional
of SRY converge on NRA, which directly binds to coactivator or repressor that is expressed early on (E)
the SRY promoter (, ). NRA (more com- in the mouse AGP (). During adrenal development
monly known as SF) is one of the most essential CITED and WT cooperatively upregulate Nra in
factors of gonadal and adrenal development, and the primordial adrenal gland (). Cited-deficient
NRA mutations account for one fifth of the cases of male mice display delayed testis determination and
,XY gonadal dysgenesis (). NRA is a master distorted testis histology (). Although it is likely
regulator of gene expression throughout gonadal and that a CITED/WT/NRA-dependent mechanism
adrenal development, in steroidogenic tissues, and operates in the gonadal part of AGP, too, CITED is at
during gametogenesis (). NRA mutations are best a relatively weak indirect inducer of Sry (, ).
associated with a spectrum of phenotypes: from ,XY There are no known cases of human DSD associated
gonadal dysgenesis and female external genitalia to with SIX, SIX, LHX, or CITED loss of function
normal male sexual development with distal hypo- (). A recent report speculates that a related gene,
spadias (, ). NRA is highly expressed in all CITED, might be more relevant than CITED in

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 871


REVIEW

humans (). CITED is upregulated after sex de- duplication of the genomic area harboring the human
termination in the fetal testis, whereas CITED is DAX gene (Xp) display a range of phenotypes from
ubiquitously expressed in  to  wpc human testis partial to complete gonadal dysgenesis, emphasizing the
(). However, there are no data available from gonads DAX dosage sensitivity of male sex determination (,
at the bipotential stage, and therefore the involvement ).
of CITED/ in human testis determination remains to Male-to-female sex reversal in Dax-overexpressing
be shown. mice is likely due to functional antagonism between
SRY/NRA and DAX (). Excess DAX in-
Module 3 terferes with activation of SRY downstream targets,
At the center of module  is WT, a TF that regulates most notably Sox, by inhibiting the binding of Sox
not only the expression of Sry but also that of Nra, stimulatory factors (SRY and NRA) to the Sox
Dax, and Amh. Owing to its direct (SRY is a direct promoter region (, ). Although there are no
target of WT) and indirect (via upregulating other known definitive reported cases of XY sex reversal
SRY expression-stimulating factors) effects, WT is due to DAX loss of function, Dax-deficient mice

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


the most important regulator of SRY expression in display a variety of gonadal phenotypes from
humans (, ). complete male-to-female sex reversal to slightly
There are many isoforms of WT that display distorted prenatal testis development depending on
differential transcription regulatory functions, but the their genetic background (–). Similar to
main difference between the major isoforms is the overexpressing mice, disorder of male sex de-
presence or absence of a three–amino acid sequence termination stems from reduced expression/action
[lysine-threonine-serine (KTS)]. In mice, an isoform of SOX (, ). Interestingly, DAX expression
that lacks the KTS sequence, that is, WT(2KTS), but is induced in the human ,XY indifferent gonadal
not the longer isoform, that is, WT(1KTS), has been primordium very early and roughly  week before
shown to regulate the expression of Nra together SRY, whereas in mice Dax is detected after Sry, at
with LHX, making WT(2KTS) part of module  E. (). Whereas DAX expression is main-
(). This is an example of at least somewhat dif- tained in both sexes through sex determination and
ferential functions that the isoforms have in devel- in the fetal gonads in humans, in mice the expression
opment and tissue maintenance as also illustrated by in post–sex determination gonads is sexually di-
phenotypic dissimilarities between Wt(2KTS) and morphic with increased expression in the ovaries
Wt(1KTS) knockout mice (). Wt(2KTS)-null (, , ). These data suggest that DAX might
mice have streak gonads in both sexes and abnormal play a somewhat different role in sex determination
Müllerian or Wolffian duct development, whereas and the maintenance of gonadal function in humans
Wt(1KTS)-deficient mice display XY sex reversal and mice. Although it is unlikely that DAX would be
(). These data suggest that WT(2KTS) plays the crucial for Sry regulation in mice owing to temporal
primary role in GR development, and once the disconnect of its expression, it might be involved in
bipotential gonad is formed, WT(1KTS) induces a the same process in humans. Nonetheless, for male
testis differentiation cascade in XY individuals. Ad- sex determination to succeed its dosage needs to be
ditional pieces of data imply that the situation might balanced in both species. Moreover, the action of
be more complicated in practice, and WT(1KTS) DAX during gonadal development might vary
has been implicated in testis determination via post- depending on the availability of corepressors or other
transcriptional regulation of Sry mRNA (). interacting proteins ().
WT(2KTS) strongly activates the SRY promoter in
humans and generally displays stronger control on other Module 4
promoters [AMH and androgen receptor (AR)], as well Whereas the three previous modules centered on
(, ). In vitro data show that human WT directly individual TFs, their upstream regulatory mechanisms,
binds to the SRY promoter and induces its transcription. and multilayered interaction between the modules,
Interestingly, WT and SRY then synergistically regulate module  is different in nature, as it is needed to make
gene expression in the nascent testis (). Both major DNA accessible for transcriptional activators and thus
isoforms of WT have also been suggested to cooperate enable the function of modules  to .
with GATA to induce SRY in humans (). Moreover, DNA methylation status of a gene promoter
WT interacts with NRA to regulate SRY, but also region is considered an indication of its transcrip-
AMH and DAX (). Involvement of DAX brings tional activity. Whereas hypermethylated promoters
another layer of complexity, as DAX has been shown are associated with silent genes, actively tran-
to impinge on WT/NRA function by physically scribed genes are usually characterized by promoter
opposing their interaction. Dosage of DAX is appar- hypomethylation. The Sry promoter region is hyper-
ently also important because DAX overexpression has methylated in the mouse embryo at E., but this
been associated with XY sex reversal both in humans genomic region undergoes spatially gonad-restricted
and mice (–). ,XY patients who carry a demethylation in the XY embryo within the

872 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

subsequent days and becomes hypomethylated by and indirect data imply that all of the modules are
E. (). Paralleling Sry downregulation, the sites activated to a similar extent in XX and XY embryos
are remethylated by E.. These stage-specific prior to E., which is consistent with the notion that
demethylation/remethylation events at the Sry pro- all of the cells within the gonadal primordium are
moter are undoubtedly critical for proper expression poised between two developmental pathways. The
of Sry, but the underlying mechanisms are yet to be decision as to which path to follow depends on the
identified. An obvious candidate with demethyl- presence (endogenous or introduced) or absence of
ase activity and association with Sry regulation, the Sry gene that then dictates the fate of the gonad by
GADDg might not play a role, as the Sry promoter inducing differentiation of pre-Sertoli cells ().
is hypomethylated in Gaddg-null mice at E. Consistently, the gene expression patterns in pre-
(). DNA methylation has been shown to control sumptive testes and ovaries bifurcate soon after the
SRY in human cell lines, and SRY expression is onset of SRY expression. The ovarian pathway and
stimulated by a DNA-demethylating agent (). differentiation into granulosa cells, the female somatic
Histone modifications are also an elemental part counterpart, is concomitantly suppressed through

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


of a genome’s epigenetic regulation. Some histone downregulation of Wnt by FGF, an indirect target of
marks are associated with repressed transcriptional SRY (, ).
status, whereas others are found at loci of active
transcription. Methylation of histone  at lysine  Sertoli cell specification
(HK) belongs to the former. HK demethylase, Sertoli cells are the first somatic cell type to be specified
lysine demethylase A (KDMA), has been implicated in the presumptive testis, and a sufficient number of
in Sry regulation in mice, and Kdma-mutant mice them are required to initiate and maintain testicular
exhibit reduced levels of Sry and male-to-female sex differentiation and block the ovarian pathway. Given
reversal with a variable phenotypic penetrance their critical role at the onset of male development, it is
depending on their genetic background. KDMA important to understand what determines the number
probably acts independently from other modules, as of pre-Sertoli cells, as initially defined by expression of
the expression of Gata, Nra, Wt, and others is not SRY and subsequently by that of SOX. Whether the
altered in Kdma-deficient mice at E. (). Sertoli cells of an adult individual are the descendants
According to a model proposed by Kuroki et al. (), of the cells that activated Sry during its narrow window “Sertoli cells coordinate the
KDMA accumulates at the Sry locus in the XY of embryonic expression, or whether it is possible to differentiation of all other cell
bipotential gonad prior to induction of Sry. This re- recruit new cells into Sertoli cell fate later, has been types within the testis,
sults in HK demethylation, deheterochromatiniza- debated (, ). Using a lineage-tracing approach, including germ cells.”
tion, and opening up of the chromatin structure, Sekido et al. () concluded that SRY acts uniquely
allowing access of TFs. However, based on a recent within the supporting cell lineage to direct the cell fate
study, removal of repressive marks is not sufficient but decision, and under normal conditions cells that have
H lysine  acetylation, a positive histone mark, is not initially expressed SRY are not recruited into this
also required for the proper activation of Sry ex- lineage. According to this hypothesis, new Sertoli cells
pression (). What is responsible for recruitment of cannot be recruited from Sry-negative somatic cells of
histone-modifying enzymes to the Sry promoter at the the gonadal primordium or the surrounding mesen-
correct time is not known. chyme. Svingen and Koopman (), however, pro-
vided many lines of evidence supporting the notion
Over the threshold that mesonephric, CE, or GR cells that express Sox
According to the presented model (Fig. c), these four at a low or undetectable level can be persuaded to
modules constitute the molecular machinery that gov- become SOX-positive pre-Sertoli cells. Furthermore,
erns the regulation of mouse Sry. Species-dependent research performed on chimeric mice shows that XX
differences in the function, significance, regulation, and somatic cells, which by definition are Sry-negative, can
even the identity of the modules are likely, and the adopt Sertoli cell identity (). These data strongly
current knowledge does not allow any comprehensive support the idea of cell-nonautonomous entry into
conclusions about SRY regulation in humans to be the Sertoli cell fate. There is also a dramatic FGF-
drawn. However, gene knockout studies in mice and stimulated increase in the number of pre-Sertoli cells
naturally occurring mutations in human have indicated between E. and E. as SRY/SOX-expressing
that induction of SRY depends on flawless concerted cells proliferate, resulting in an overall increase in
action of different modules, and failure of any of them the XY gonad size (–). In Wt(1KTS)-deficient
might result in subthreshold expression of SRY and and Six/-mutant mice, cell proliferation within this
impaired male sex determination and differentiation. period is reduced, resulting in too few Sertoli cells to
Once the threshold level of SRY protein is reached, maintain testicular differentiation and consequent
downstream events are triggered, and SRY alone or in male-to-female sex reversal (, ). Notably, it is
synergy with other TFs (such as WT) consequently not just the level of Sry expression per cell, but also the
stimulates the expression of male-specific genes. Direct number of Sry-expressing pre-Sertoli cells that is

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 873


REVIEW

critical for entry into the testis development pathway maintenance of Sertoli cell differentiation and identity,
(). suppression of the ovarian pathway, maintenance
The specification of Sertoli cells is a key event of the Wolffian structures, and repression of the
during testis formation (Fig. a). Sertoli cells co- Müllerian ones.
ordinate the differentiation of all other cell types The expression of mouse Sox is controlled by
within the testis, including the germ cells. Having a factors that bind to a .-kb enhancer region, called
single cell type as the orchestrator of testis differen- testis-specific enhancer of Sox (TES) (). Prior to
tiation allows the fate of several cell lineages to be SRY, NRA binds to and activates TES, then from
coordinately specified. E. on SRY and NRA synergistically upregulate
Sox expression via TES, and after downregulation of
SOX9 SRY, Sox expression is maintained by FGF, pros-
Commencing at ~E. in mice, upregulation of Sox taglandin D synthase (PTGDS), doublesex and mab-
by SRY is a critical step for the development of a –related TF (DMRT), and, as described above, by
fertile male (Figs. a and a). Similar to Sry, Sox is SOX itself (Fig. a) (, ). The expression of Fgf

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


upregulated in the GR following a center-to-pole is directly or indirectly stimulated by SOX, and these
pattern (). Intriguingly, the successive waves of two thus seem to form a positive feedback loop that
Sry and Sox expression are not induced by a GR maintains the high expression of both the genes and
central domain–derived diffusible factor, as they still represses that of Wnt, a key gene in ovarian devel-
occur in isolated and cultured anterior and posterior opment ().
pole GR segments (). In ,XY human embryo, Interestingly, an FGF signal radiating outward
SOX is upregulated following SRY, and it reaches from the center of GR is needed for the sustained
the highest expression at  dpc (). SOX protein expression of SOX at the anterior and posterior poles
is not detected in a -wpc human XY embryo, but a (). Both FGF and prostaglandin D (PGD, a
high level of SOX is observed in -wpc fetal human product of PTGDS) have been implicated in re-
testis (). SOX is indispensable and sufficient for cruitment of SOX-low/negative cells into Sertoli cell
testicular organogenesis, and SOX can functionally fate as a means of increasing the population size of pre-
substitute for the lack of SRY and induce testis de- Sertoli cells, which is critical for testis formation
velopment in an XX embryo both in humans and (–). Emphasizing the importance of FGF
mice (–, –). signaling, Sertoli cell differentiation is impaired in Fgf
The action of SOX during human sex de- and FGF receptor (Fgfr) (FGF receptor)–null mice,
termination is strictly dose sensitive, and a critical and they consequently exhibit partial or complete
threshold for SOX expression has to be reached to male-to-female sex reversal (, , ). The sig-
maintain testicular differentiation. Notably, hetero- nificance of FGF/FGFR signaling in the nascent
zygous mutations of SOX are associated with com- human testis has not been characterized, but a het-
plete ,XY gonadal dysgenesis, whereas duplication of erozygous point mutation in FGFR has recently been
the region in chromosome  that houses the human associated with ,XY male-to-female sex reversal
SOX gene results in ,XX testicular DSD (, , (). Additionally, FGF is expressed very early
, ). SOX duplication is, however, a relatively ( wpc) in the human embryonic testis, suggesting a
rare cause of ,XX testicular DSD, but there are a role during human testis differentiation, too ().
number of reports of similar cases with an identified SOX also forms another feed-forward loop: PTGDS
mutation in the putative SOX regulatory region, is a direct target of SOX, and PTGDS in turn pro-
suggesting that ectopic and aberrant activation of motes SOX transcriptional activity by promoting its
SOX might be the causative factor there (–). nuclear translocation (, , ). The significance
Similarly, mutations at the SOX upstream enhancer of PTGDS for maintenance of SOX expression is
regions can result in partial or complete loss of SOX probably less important than that of FGF signaling, as
transcription in the ,XY embryo and development testicular organogenesis is only mildly affected in
of an ovary instead of a testis (). The critical Ptgds-deficient mice and no known cases of DSD
thresholds for gene activation seem to be species- with a mutated PTGDS gene in human patients have
dependent, as Sox heterozygous mutant males are been reported (, ). Working as an autocrine and
normal and a homozygous deletion is required to paracrine factor, PGD is, however, a potent agent,
reverse the sex of the affected XY mice (). and it can partially masculinize female embryos in its
These findings make SOX the most important own right (, ). DMRT, a gene needed for the
direct target of SRY. Once induced above a certain maintenance of Sertoli cell or male identity, supports
threshold level, SOX then orchestrates a cascade of the expression of Sox and represses the female
gene interactions and maintains its own expression via pathway by suppressing forkhead box L (Foxl) ().
an autoregulatory mechanism (Fig. a) (). SOX The significance of TES for human SOX expression
coordinates the function of a male-specific tran- has been justifiably questioned, as there are no known
scriptional network to achieve the following outcomes: cases of DSD associated with TES mutation in

874 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

humans. It has rather been shown that human SOX is XY double-knockout gonads SRY was shown to ac-
regulated by a distal enhancer element, termed RevSex tivate the expression of  genes, including desert
(). hedgehog (Dhh), Sox, and cytochrome P family
 subfamily B member  (Cypb) (). DHH is
AMH crucial for fetal Leydig cell specification, SOX is a
One of the most critical factors secreted by nascent close homolog of SOX and might compensate for its
Sertoli cells is AMH. It belongs to the TGFb family of loss, and retinoic acid (RA)–degrading enzyme
growth factors and its production is maintained in the CYPB is specifically expressed in the testis to
testis until puberty (). For this reason it is prevent premature meiotic entry of germ cells (,
considered a specific marker for Sertoli cell functional , ). These data provide a more multifaceted
immaturity. AMH has a pivotal role in driving the
regression of Müllerian ducts, whereas androgens
derived from the fetal Leydig cells are responsible for
the maintenance of Wolffian ducts and their sub-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


sequent development into male accessory sex glands
and urogenital structures. Recently, AMH has been
proposed to control gonocyte entry into a mitotically
inactive state during fetal development, and in the
neonatal mouse AMH has been implicated to promote
germ cell transition from gonocytes to spermatogonia
(, , ). When AMH or its action is missing,
Müllerian derivatives persist, emphasizing the signif-
icance of AMH for male urogenital tract development
().
AMH is upregulated in the human embryonic
testis during the eighth developmental week, and a
high protein expression of AMH is detected in the
human fetal testis from  wpc on (, ). Similar to
Sry, Amh expression is regulated by a number of
essential testicular factors: SOX, NRA, WT, and
GATA all have a stimulatory effect on Amh ex-
pression, whereas DAX inhibits its transcription via
an antagonistic function (–). Amh is a direct
target of NRA, but NRA also regulates Amh
expression synergistically with WT(2KTS) and
SOX// (, ). A similar synergism has been
observed between GATA and WT(2KTS) ().
These are just a few examples of the complex network
of factors and interactions behind the regulation of
Amh transcription. Although SOX is needed for the
induction of Amh expression, it remains to be elu-
cidated how different factors converge on Amh reg-
ulation and how they collaborate, cooperate, and
interact to control the sex-dependent expression of
AMH ().

Other SOX family genes


For good reason, SOX has been regarded as a de facto
Figure 4. (a) Regulation of SOX9 and its role in testiculogenesis. Sox9 is initially induced by
inducer of male development that is needed for both the synergistic action of SRY and NR5A1. Later, SOX9 maintains its own expression either directly
activation of a testis-specific transcriptional program or by feed-forward loops with PTGDS, FGF9/FGFR2, and DMRT1. SOX9 orchestrates testicular
and repression of the ovarian pathway (Fig. a). development by maintaining Sertoli cell differentiation/identity and repression of the ovarian
However, Nicol and Yao () asked a very important pathway either directly or via DMRT1 and FGF9 signaling. DHH and AMH are among the key SOX9
question: What is to become of the bipotential gonad downstream genes needed for the successful development of the male urogenital system. (b)
Maintenance of the testicular phenotype. DMRT1 maintains male sex postnatally together with
if both SOX and b-catenin, SOX’s functional
SOX9. Besides maintaining the expression of testicular genes, they repress those that are active in
counterpart in the female, are absent? Interestingly, ovarian development (Wnt4, b-catenin). DMRT1 also silences RA action at sex-sensitive loci (Foxl2,
both XY and XX gonads progressively lean toward the Esr2). When DMRT1 is absent, RA is able to activate feminizing genes, which also repress Dmrt1 and
testis fate, but XY double-knockout gonads are more Sox9 that are needed to maintain Sertoli cell identity. AR signaling impinges on Foxl2 transcription
masculinized than are their XX counterparts. In the in mouse Sertoli cells. RAR, RA receptor; T, testosterone.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 875


REVIEW

role for SRY in testis determination than previously connected at their proximal regions to a single cord-
thought. like structure, the presumptive rete testis that runs
In general, the role of SOX family genes in testis parallel to the mesonephros. The individual cords
determination is intriguing. Besides the two indis- further elongate and start to coil by E., which is
pensable genes (SRY and SOX), other members of probably an indication that the intratesticular space
the family have been implicated in transformation of becomes limiting due to emergence of the tunica
the bipotential gonad into a testis. Following SOX, albuginea, a stiff fibrous capsule surrounding the
SOX and SOX are upregulated in the mouse testis (, ).
XY gonad at ~E (, ). Sox-null mice display SOX has been shown to be indispensable for testis
progressive degeneration of the seminiferous epi- cord formation, but during subsequent testicular
thelium and infertility by  month of age (, ). differentiation its function can be substituted for by
Use of inducible knockout mouse models has shown SOX and SOX (, ). As reviewed by Svingen
that the initial testis determination is dependent on and Koopman (), in vitro and in vivo data imply
SOX, but SOX and SOX act synergistically dur- that neurotrophic tyrosine kinase receptors and their

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


ing differentiation and maturation of testicular ligands are involved in the aggregation and cord
structures (, ). SOX and SOX have both formation process of pre-Sertoli cells. FGF has also
been associated with female-to-male sex reversal in been implicated in testis cord formation but its effect
overexpressing transgenic mouse models and in might be indirect (stimulation of pre-Sertoli cell
,XX patients with a duplication in the genetic proliferation and maintenance of their identity) (,
regions that encompasses either of these genes (, ). Inhibition of TGFb, Activin, and Nodal signaling
, , ). The ability of SOX and SOX to also impinges on early testiculogenesis, suggesting that
direct male development can be explained by two ligands and receptors of these pathways are needed for
different mechanisms: either they substitute for lack normal male gonadal development (). Notably,
of SRY, or they activate the same genes as SOX. germ cell–derived molecules are not essential for cord
There is evidence for both (, ). A lack of formation, as testis cords do develop in germ cell–
gonadal phenotype in prenatal Sox- and Sox- deficient mice (, ). However, according to a
deficient mice suggests that neither is essential for recent report, cord formation is severely affected in
testis determination or differentiation. Recently, SOX We/We (Kit mutant) mice that have very few intra-
C group proteins SOX, SOX, and SOX have gonadal germ cells due to a PGC migratory defect
also been implicated in testicular organogenesis (). The authors take it as a proof of importance of
(). Sox deficiency results in elongated gonads in bidirectional (Sertoli–germ cell) signaling during early
both sexes at E. and in more testis cords in XY testiculogenesis, but further independent approaches
embryos (). In conclusion, SOX family genes are are needed to confirm that the phenotype is not due to
absolutely crucial for testicular organogenesis. Their indirect effects of abnormal SCF/KIT signaling.
functional redundancy due to shared transcriptional
targets calls for their levels of expression to be tightly Origin of other testicular cell types
regulated in both sexes, as misregulation might result As discussed above, hPGCs arrive at the developing
in impaired gonadal development and partial or gonad over a wide temporal window, and many
complete gonadal dysgenesis. hPGCs are found outside the presumptive testis at
the time of SOX induction during the seventh week
Formation of the fetal testis of development (, , , ). It is not known
when cord formation and tubularization have pro-
Testis cord formation ceeded far enough in the human fetal testis to im-
Soon after the emergence of SOX-expressing pre- pede incorporation of late migrating PGCs into
Sertoli cells, cells within the long and thin GRs start the testis cords. It is, however, conceivable that the
to reorganize. Aggregation of pre-Sertoli cells initi- formation of the tunica albuginea makes testicular
ates testis cord formation, and germ cells are sub- entrance for PGCs, or any other cell type, very
sequently encased within the nascent cords whereas difficult if not impossible (). Formation of the
non-Sertoli somatic cells are left on the outside (Fig. testis cords brings changes to the nomenclature and
a). The gonadal length in mice is reduced until the properties of the cell types involved: pre-Sertoli
E., whereas its width quadruples from E. to cells become fetal or immature Sertoli cells that
E. (). Around  protocords are initially undergo a transition from a mesenchymal cell-like
formed but their number is reduced to ~ by E. state to a polarized epithelial cell, and PGCs that
when these structures form sharp-edged loop-like are engulfed by testis cords become gonocytes (or
structures that are surrounded by the testicular prespermatogonia/prospermatogonia in rodents)
interstitium, and are now referred to as testis cords whose fetal and prepubertal development is the
(). As a result of Sertoli cell proliferation, the testis topic of one of the following sections. PGC-to-
cords subsequently become longer and wider and are gonocyte transition encompasses a significant shift

876 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

in germ cell transcriptome toward what could be (–). Although being implicated in control of
called a gonadal-stage germline marker expression SSC maintenance and differentiation, the ultimate
profile. function for these cells is yet to be defined. Recently, it
Not only the Sertoli and germ cells but also all the was shown that the ontogeny of testicular macrophage
other cell types present in the adult testis originate subsets differs, and although the interstitial macro-
from precursors that are specified or recruited into phages are derived from the yolk sac, the origin of
testis parenchyma during fetal development. VECs and peritubular macrophages is distinct and they emerge
peritubular myoid cells (PMCs) are intimately involved postnatally from bone marrow–derived progenitors
in the formation of testis cords. VECs are derived from (). As the mouse ages, the interstitial compartment
the mesonephros. Vascular endothelial growth factor is also replenished by bone marrow–derived
(VEGF) and platelet-derived growth factor (PDGF) progenitors.
signaling mediate the migration of VECs to the XY Development of Wolffian derivatives and external
gonad from E., and formation of testicular arteries genitalia depends on production and secretion of
is subsequently observed (–) (Fig. a). For- androgens by fetal Leydig cells relatively early during

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


mation of testicular vasculature and lymphangio- fetal development. Upregulation of genes encoding
genesis has been recently reviewed elsewhere (). the key known components of testicular steroido-
Blocking the migration of these cells is detrimental genesis [LHCGR, STAR, HSDB, hydroxysteroid
for testicular morphogenesis, and endothelial cells b-dehydrogenase  (HSDB), CYPA, and
have a crucial role in establishing tissue architecture CYPA] is observed already roughly a week after
during testiculogenesis (–). Platelet and en- SRY, during the eighth wpc, and they generally
dothelial cell adhesion molecule  (PECAM)–positive reach a plateau of high expression by the end of the
putative VECs are first observed in the human testis at following week (). Similar to PMCs, the embry-
 wpc, but vascularization proceeds slowly and large onic origin of (fetal) Leydig cells is a topic of heated
vessels with PECAM-stained endothelia are first seen debate. Disruption of embryonic Hedgehog (HH)
in  wpc testicular interstitium, but they might begin signaling results in abnormalities in both the cell
to form as early as  wpc (). populations, suggesting that HH signaling might act
Despite decades of research, the origin of PMCs on a common precursor. The testis-specific member
remains profoundly mysterious. Similar to VECs, they of the HH family of secreted proteins, DHH, pro-
“…SOX family genes are
were previously thought to derive from the meso- duced by (pre)Sertoli cells, is one of the first genes to absolutely crucial for testicular
nephros, but more recent research has elucidated that be expressed in the mouse XY gonad after Sry (). organogenesis.”
the immigrant cells are exclusively endothelial and do In the mouse testis, HH signaling plays a crucial role
not contribute to PMCs or other interstitial lineages in specification and differentiation of fetal Leydig
(, –). PMCs at the periphery of mouse testis cells and PMCs (, –). Whereas Dhh-mu-
cords become morphologically distinct by E. (). tant mice are azoospermic and display abnormal
Clearly identifiable testis cords or protocords in  wpc testicular histology, DHH mutations in humans have
human fetal testis seem to lack PMCs, but smooth been associated with complete or partial ,XY go-
muscle actin–positive PMCs enclose the testis cords by nadal dysgenesis (, –). In conclusion, both
 wpc (). Leydig cells and PMCs are likely specified by Sertoli
One remarkable difference in the histology of cell–dependent mechanisms. This signifies the need
rodent and human testicular tissue is the organization to maximize the population size of pre-Sertoli cells
of peritubular cells. Whereas only one layer of PMCs by direct (high endogenous SRY) or indirect (re-
encapsulates rodent seminiferous tubules, the sheet of cruitment into Sertoli cell fate via secreted factors)
peritubular cells in humans is thick and consists of five means. Specification of the interstitial cell lines, in-
to seven cellular layers and intertwined ECM, and is cluding fetal Leydig cell population and the origin of
called the lamina propria. Besides PMCs, fibroblasts adult-type Leydig cells, have been recently reviewed
are also incorporated into the human lamina propria. elsewhere ().
Impaired spermatogenesis is often associated with
thickening of the lamina propria due to accumulation
of ECM between the cellular layers (). Both Sertoli Testicular Descent
cells and PMCs contribute to the ECM and the basal
lamina of the seminiferous epithelium (). Testicular descent from intra-abdominal position into
An intriguing population of testicular somatic cells the scrotum is a continuum that has often been de-
that is often neglected is the macrophages. Although it scribed to have two main phases: the transabdominal
has been known already for quite some time that phase and the inguinoscrotal phase. In the trans-
interstitial macrophages intimately interact with abdominal phase the testes are anchored close to the
Leydig cells and play a role in androgen production, a inner entrance of the future inguinal canal, and in the
new subset of macrophages was recently discov- inguinoscrotal phase the testes pass through the in-
ered intermingled with PMCs in the mouse testis guinal canal into the scrotum. The two phases have

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 877


REVIEW

been described to occur in humans between the th inguinal canal is plugged by a fat pad on the epi-
and th gestational weeks (GWs) and between the didymis ().
th and th GWs, respectively (, ). In rodents, The inguinal canal differentiates around the
the inguinoscrotal phase occurs postnatally around gubernaculum during early fetal development in
puberty (, ). humans (). Prior to the transinguinal passage of the
Gubernaculum is a mesenchymal structure be- testes, swelling of the gubernaculums has been ob-
tween testis and inguinal area and it has an important served and this widens the inguinal canals (, ).
role in testicular descent. According to mouse models, However, no eversion of the gubernaculum occurs in
the transabdominal phase of testicular descent is de- the inguinoscrotal phase of testicular descent in
pendent on Leydig cell hormone insulin-like peptide  humans (). After the testes, epididymides, and
(INSL) inducing male-like development of the gubernaculums have passed through the inguinal
gubernaculums (the shortening of the cord and en- canal as a unit, possibly with the help of intra-
largement of the bulb) (–), which anchor the abdominal pressure, the gubernaculums start to
testes into the inguinal area and prevent them from shrink (, ). The testes and epididymis and

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


ascending when the fetus grows (). Signaling of gubernaculum migrate from the external inguinal ring
INSL and its receptor [relaxin family peptide receptor into the scrotum following the genital branch of the
 (RXFP)] affects myogenic differentiation of the genitofemoral nerve (). The transinguinal descent
gubernacular mesenchymal cells possibly via WNT/ of testes starts around GW  to , and usually testes
b-catenin pathways (, ). If the male-like de- have descended into the scrotum by the th GW
velopment of the gubernaculum is disrupted, the testes (–). The scrotal ligament remains as a remnant
are freely moving and they locate high in the abdomen of gubernaculum and the processus vaginalis closes
(–). ().
In humans, both male and female fetuses have been Androgens are important for the inguinoscrotal
described to show in early pregnancy an inner descent phase of testicular descent, and this phase is usually
of gonads that is dependent on descending anlage of disrupted both in mice and boys with androgen in-
the diaphragm (). Later on, after regression of the sensitivity (testicular feminization syndrome) (,
Müllerian ducts and the involution of the cranial ). However, mutations of the AR are rare in pa-
mesonephric ligament in male fetuses, the testes glide tients with isolated undescended testis (). In the
across the genital ducts (Wolffian ducts) and dive into mouse, androgens also cause the regression of the
swollen gubernaculums (). However, it has also cranial gonadal ligament, the cranial suspensory
been suggested that gubernaculum is attached only to ligament, during the transabdominal phase of tes-
ductus deferens and epididymis but not directly to ticular descent (). However, androgens are
testis (, ). INSL is likely to play a role in needed already before the inguinoscrotal phase, and
testicular descent also in humans, because it has been only exposure to antiandrogens during the out-
detected only in amniotic fluid of male fetuses (, growth of the gubernaculum or earlier disrupts the
). Congenital cryptorchidism has been associated postnatal inguinoscrotal descent of testes in rodents
with reduced INSL levels in cord blood (, ). (, ). The target of androgens in testicular
However, mutations in the genes of INSL or its descent is unknown, but animal studies suggest that
receptor are rare in cryptorchid patients (, ). androgens affect migration of the gubernaculum in
Boys with persistent Müllerian duct syndrome due to the inguinoscrotal phase indirectly via genito-
mutations in the genes of AMH or its receptor femoral nerve and its neurotransmitter calcitonin
have abnormally long gubernaculums and unilateral gene–related peptide (CGRP) and inguinoscrotal fat
cryptorchidism or bilateral intra-abdominal testes pad (, ). In addition to androgens, the INSL/
(, ). Thus, also AMH is likely to have a role in RXFP-mediated pathway has also been suggested
the transabdominal phase of testicular descent in to have a role in the inguinoscrotal descent of the
humans, in the shortening of the gubernacular cord. testis ().
In the mouse, AMH is not needed for the descent of The prevalence of undescended testis has been
the testes (). However, it has been suggested that reported to be % to % among full-term boys in
AMH may still affect the length of the gubernacular prospective cohort studies. Higher prevalence has
cord (). been observed among premature babies and in boys
In rodents, in the inguinoscrotal phase, the bulb of born small for gestational age (). Usually the
the gubernaculum remodels and everts into a hollow inguinoscrotal phase of testicular descent is dis-
cone, and this eversion creates the processus vaginalis rupted (). Spontaneous descent of undescended
(a diverticulum of the peritoneal membrane) (). testis is often noticed during the first few months of
Thereafter, the gubernaculum elongates to the life, likely due to temporary postnatal activation of the
scrotum similar to a limb bud and becomes sec- hypothalamus–pituitary–gonadal (HPG) axis and as-
ondarily attached to the scrotal skin (, ). The sociated increase in reproductive hormone levels (,
processus vaginalis remains open in rodents and the –).

878 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Disorders of Testicular Development including DMRT can cause ,XY gonadal dysgenesis
(). Sertoli cells are rapidly proliferating first until
Most DSDs have an unknown etiology, although our GW , then after birth during the minipuberty for the
knowledge on the molecular genetic basis of gonadal first  months of life, and finally at early puberty before
development has rapidly increased. Disorders of tes- the onset of spermatogenesis ().
ticular development can originate on different levels: Sertoli cells form the seminiferous cords that also
development of the bipotential gonad, differentiation encompass the germ cells that have migrated from the
of the testis, or function of the testis. Germ cells are not yolk sac via hindgut to the gonad. Sertoli cell–derived
necessary for gonadal differentiation that occurs under AMH induces regression of paramesonephric struc-
endocrine, paracrine, and autocrine regulation. A tures that otherwise develop to oviducts, uterus, and
bipotential gonad develops from the gonadal pri- the upper third of vagina. Mutations in AMH or its
mordium in the CE on the ventral side of the me- receptors cause the persistent Müllerian duct syn-
sonephros. Several genes are active at this early phase drome where a boy has both male and female internal
on GW  to , including GATA, WT, and the sex organs.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


nuclear receptor SF (NRA) (). Some mutations In the interstitial space some of the progenitor cells
in WT lead to ,XY gonadal dysgenesis (), differentiate to Leydig cells. These cells activate Notch
whereas some NRA mutations can cause agenesis of signaling, downregulate WT, and upregulate SF and
both the adrenal gland and gonads (). However, the steroidogenic enzymes, leading to active steroid syn-
phenotypic variation is large and adrenal agenesis is thesis. A balance between proliferating progenitor cells
not as frequent as gonadal dysgenesis (). and differentiating steroidogenic cells has to be
Differentiation of the supporting cells in the maintained. Aristaless-related homeobox (ARX) and
bipotential gonad to testicular Sertoli cells is the de- chicken ovalbumin upstream promoter TFII (COUP-
cisive step in sex-specific development. This is initiated TFII, officially NRF) are hallmarks of progenitor-
by the SRY gene that is activated on  dpc (, ). like cells, and their expression declines whereas that of
Factors that are active in the early phase of devel- SF and steroidogenic enzymes increases. Mutations
opment (GATA, SF, WT) and MAPK pathway are in ARX can lead to poor masculinization of the ,XY
controlling SRY, as well as epigenetic modulation by fetus due to a small number of Leydig cells ().
DNA methylation, histone modification, and chro- Notch signaling controls the population of the pro-
“Differentiation of…testicular
matin modeling (). Gene variants of CREB-binding genitor cells, and the loss of action leads to an in- Sertoli cells is the decisive step
protein and its related paralog p that operate in creased mass of fetal Leydig cells and a declined in sex-specific development.”
histone modifications have been associated with DSDs progenitor pool (). Mutations in the mastermind-
in some cases (). Mutations in SRY are causing like domain containing  (MAMLD) gene, a gene
,XY gonadal dysgenesis, whereas most ,XX tes- required in Notch signaling, can lead to complete
ticular DSD is caused by a translocation of SRY- ,XY gonadal dysgenesis (). Sertoli cells play an
containing piece of chromosome Y (). important role in proliferation and differentiation of
Specification of Sertoli cell lineage is secured by the Leydig cells by secreting DHH and PDGF (, ).
SRY target gene SOX (). Mutations in SOX cause Mutations leading to compromised DHH signaling
,XY gonadal dysgenesis and severe campomelic can also cause ,XY gonadal dysgenesis (). Fetal
dysplasia that are often lethal early in life (). Leydig cells involute after birth and adult Leydig cells
Mutations in the distal enhancer of SOX, RevSex, develop from an own progenitor pool ().
have also been associated with DSDs (). SOX acts In rodents, fetal Leydig cells are unable to syn-
in concert with DMRT, GATA, SF, and WT to thesize testosterone, and they secrete androstenedione
control the target genes necessary for testicular de- and other androgens that are considerably less potent
velopment. Once SOX gene is activated, it can than testosterone. Production of testosterone from
upregulate its own expression via FGF and PGD these precursors then takes place in Sertoli cells that
(, ) (Fig. a). Ovarian differentiation is promoted express HSDB (, ). In the human fetus,
by WNT, R-spondin  (RSPO), and b-catenin, which Leydig cells differentiate right after Sertoli cells at  to
are suppressed by SOX and FGF tilting the devel-  wpc, and the testis starts to produce increasing
opment into the male direction. When SOX activity is amounts of testosterone. Androgens are needed for
compromised, the central part of the gonad may de- masculinization of internal and external genital tract.
velop into testis, whereas the poles differentiate to the Testosterone is further converted to DHT in the target
ovarian direction, causing a rare ovotesticular dysgen- tissues by a-reductase, which is necessary for effective
esis. The SOX gene family includes other members in androgen action, because DHT is much more potent
addition to SRY and SOX, such as SOX and SOX, than testosterone. Development of the prostate and
that may also contribute to gonadal development, be- external genitalia is critically dependent on DHT. Loss
cause duplications of the SOX-containing gene region of function of a-reductase and defects in steroido-
in chromosome q is associated with ,XX gonadal genic machinery in Leydig cells result in various de-
dysgenesis (). Deletions in chromosome p grees of ,XY DSD where undermasculinization can

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 879


REVIEW

present with undescended testes, hypospadias, micro- decreasing levels in the ovary and increasing expres-
phallus, and a short anogenital distance up to completely sion in the testis ().
female external genitalia. Hypogonadotropic hypo- Mutations in the human DMRT gene have been
gonadism can cause a similar phenotype, albeit usually associated with ,XY gonadal dysgenesis and male-
less severe, because the Leydig cells need pituitary go- to-female sex reversal (–). Interestingly, Dmrt-
nadotropin stimulation, particularly during the latter null XY mice display overtly normal embryonic
half of pregnancy when placental chorion gonadotropin testicular development, but the Sertoli cells of these
levels decline. Inactivating luteinizing hormone (LH) mice fail to mature and do not cease to proliferate
receptor mutations lead to ,XY DSD with a female (). Germ cell maintenance in these mice is severely
phenotype (). disrupted, and they are totally depleted by  weeks of
The most common reason for ,XY DSD is in- age (, ). Adult Dmrt-null mice are charac-
sensitivity of the AR, which may lead to completely terized by dysgenic testes with few poorly organized
female external phenotype in the case of complete tubules lacking the lumen, as well as testicular fibrosis
androgen insensitivity or undermasculinization when and/or Leydig cell hyperplasia (). Cell type–specific

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


the defect is partial. Mutations are found usually in the ablation studies of Dmrt further demonstrate that
exomes of AR, but intronic variants have also been Dmrt expression in germ cells is needed for their
identified (). homing from the tubular lumen to the basement
membrane of the seminiferous epithelium, a critical
step for the onset of spermatogenesis and formation
Maintenance of Testicular Phenotype of germline stem cell pool (). Germ cell survival,
maintenance, and differentiation also depend on
The genetic sex of an individual is determined at the Dmrt expression in Sertoli cells, suggesting that
moment of fertilization. For the first  weeks the DMRT is a master regulator of Sertoli cell function.
development of XY and XX embryos is synchronized, According to these data, there might be a discrepancy
but then induction of SRY sets the ball rolling and the in the role of DMRT in testis development in hu-
future males embark on a developmental trajectory mans and mice, and DMRT is probably needed at
that ultimately gives rise to all the characteristics that an earlier stage in humans to maintain the gonadal
are collectively referred to as “maleness.” As discussed somatic cell identity.
above, determination of the gonadal phenotype de-
pends on a concerted action of a number of genes, very FOXL2 in ovarian development
few of which are critical, and the process is strictly FOXL is a critical gene in ovarian development and
regulated in time and space. Surprisingly, the sex maintenance of the ovarian function. It is expressed in
identity of gonadal supporting cells is not fixed once both granulosa and theca cells. However, the signifi-
specified but needs to be constantly maintained. cance of FOXL varies considerably depending on the
Failure to do so might result in cellular trans- species. Although it is indispensable for female de-
differentiation and compromise the reproductive velopment in the goat, and XX goat embryos lacking
function (, ). Emerging data suggest that there Foxl develop testes, in humans and mice loss of its
are two TFs that are responsible for the maintenance function is associated with ovarian failure and in-
of the sex identities of gonadal supporting cells by fertility (–). A conditional deletion of Foxl in
promoting a specific sex-dependent transcriptional the adult mouse ovary leads to reprogramming of the
circuitry and repressing the opposite one (Fig. b). ovarian granulosa and theca cells to the testicular
These two mutually antagonistic factors are DMRT counterparts, Sertoli and Leydig cells, respectively
and FOXL. (). This transdifferentiation process is driven by
induction and activation of Dmrt and Sox in the
DMRT1 in testis development ovary. Conversely, loss of Dmrt in the adult testis
DMRT transcripts are detected alongside with SRY in leads to de-repression of Foxl, downregulation of
 to  wpc male human embryos but not in female Sox, and transformation of testicular cell types into
embryos (). During the first half of pregnancy, ovarian equivalents (, ). These data provided
DMRT is principally expressed by Sertoli cells, but the first definitive proof that somatic cell identities of
thereafter the expression in germ cells becomes gonadal-supporting and steroidogenic cells are not
dominant. However, prenatal germ cells, from -wpc irreversible, and active measures need to be taken to
PGCs to mitotically quiescent gonocytes, express maintain their characteristics and functionality. Such
DMRT (). Importantly, no DMRT expression is plasticity of cell fates is indeed surprising because one
observed in the somatic compartment of the ovary, might expect that gonadal somatic cells would
and it is lost from oogonia upon meiotic entry (, employ a set of mechanisms to permanently silence
). A similar expression pattern is found in the the genes that are involved in sex determination
mouse, and DMRT expression becomes sexually and differentiation of the opposite sex, such as
dimorphic at the onset of gonadal differentiation with chromatin and histone modifications. Self-evidently,

880 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

however, this is not the case and the gonadal phenotype of age (). These cases of testicular dysgenesis
must be considered a relatively labile property. are phenocopied by conditional inactivation of Sox
on a Sox-knockout background resulting in down-
The maintenance mechanism of gonadal somatic regulation of Dmrt (, ). In conclusion, these
cell sex identity data indicate that SOX/ are needed for the main-
How does the loss of Dmrt then mechanistically con- tained expression of Dmrt. The opposite is equally
tribute to the activation of female-specific transcrip- true, and the data suggest that SOX is involved yet in
tional circuitry and adoption of ovarian supporting cell another feed-forward loop to maintain its high ex-
identity? It has been demonstrated that fetal sex de- pression (Fig. ) ().
termination, transdifferentiation, and maintenance of Roughly a -month period is needed for the effects
the sex postnatally are mechanistically related to each of Dmrt loss to become visible independent of the
other (). DMRT maintains male sex postnatally time of the genomic rearrangement (, ). This
together with SOX, and the genes that they repress indicates that there are redundant mechanisms that act
include those that are active in the female sex de- together with DMRT to maintain testicular identity

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


termination network (Fig. b). Additionally, DMRT and buffer against detrimental effects of stochastic
silences RA action at sex-sensitive loci [e.g., Foxl, es- changes in DMRT expression. The factors that are
trogen receptor  (Esr)] (). When DMRT is absent, involved act either to maintain SOX expression or
RA is able to activate feminizing genes, which, besides repress the induction of feminizing genes. Such a
promoting the granulosa cell fate, repress the male function has been assigned at least to FGF, that,
program that is needed to maintain Sertoli cell identity. besides stabilizing SOX and stimulating its expres-
Interestingly, AR signaling also impinges on Foxl sion, is primarily responsible for repression of female
transcription in mouse Sertoli cells, suggesting that genes, including Wnt (, , ). Importantly,
testosterone is actively involved in the maintenance of note that gonadal SOX expression is repressed by
Sertoli cell identity (). Thus, two highly potent WNT and its downstream effector b-catenin, and
hormones, testosterone and RA, work in opposing di- WNT also indirectly impinges on Sry (–).
rections with respect to maintenance of testicular go- Furthermore, it is also possible that estrogen synthesis
nadal phenotype, and DMRT is required to restrict the may have to reach a specific threshold for the proper
action of RA to permit its beneficial spermatogenesis- onset of the feminizing program and repression of the “In humans, fetal and
promoting action and prevent the harmful induction testicular one. Estrogen dependency and existence of prepubertal differentiation of
of the feminizing genes (Fig. b). Interestingly, an FGF-like pro-maleness mechanism might together the germline is characterized
enforced expression of Dmrt alone in XX mouse provide an explanation for the -month schedule of by asynchronicity….”
fetal gonads is sufficient to drive testicular differ- testis-to-ovary transformation.
entiation and development of male secondary sex Based on association of human chromosome p
characteristics (, ). deletions removing DMRT with ,XY male-to-
As discussed above, DMRT is expressed in the female sex reversal, it is reasonable to assume that
GRs at the time of sex determination in both DMRT function in humans and mice is conserved.
humans and mice (, ). If the bipotential However, Dmrt-null mice are born male, whereas
gonad assumes ovarian fate, Dmrt is promptly ,XY patients, who carry a mutation at chromosome
downregulated, whereas in the testis its expression is , may have fully feminized external genitalia and
enhanced (). When does the DMRT function display complete gonadal dysgenesis (, ). The
then become essential for the maintenance of tes- difference may be ostensible and can be explained by
ticular gonadal identity? Based on the available data, considerably longer human gestation that may permit
it is hard to judge. Both global Dmrt-null mice, as reprogramming from testis to ovary already during
well as mice where Dmrt has been ablated in a fetal development. Nonetheless, species-dependent
Sertoli cell–specific manner from E. on, have differences in molecular control of the process can-
seemingly normal prenatal testis development (, not be ruled out.
, ). However, in both, the maintenance of
testicular structures is severely compromised soon
after birth. Whereas global Dmrt deficiency results Regulation of Gonocyte Proliferation,
in germ cell depletion and Sertoli cell hyperplasia by Quiescence, and Differentiation
postnatal day , the Sertoli cell–specific ablation of
Dmrt from E. on does not seem to have any A theory prevails that the sex identity of PGCs is
immediate effect on testiculogenesis until  weeks of undefined prior to determination of the somatic sex.
age, when the first signs of gradual loss of Sertoli cell The sex of PGCs and their entry into the male or
identity become evident (, ). Subsequently, female gametogenic pathway is dictated by strong
the number of SOX-expressing gonadal-supporting inductive signals from the gonadal somatic cells. In
cells is dramatically reduced, the gonadal architecture is humans, embryonic/fetal germ cells can be subdivided
rearranged, and Foxl is strongly induced by  month into three categories (in chronological order): PGCs

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 881


REVIEW

(migratory), gonocytes (proliferatively active), and fashion and over a relatively long period of time
prespermatogonia (displaying variable mitotic activ- (–). Owing to the absence of a marker that is
ity). This is confirmed by recent RNA sequencing exclusively expressed in the gonocytes, it is also very
analysis performed on germ cells isolated from - to hard to pinpoint a specific time point when gonocyte
-wpc human male embryos, indicating that there are proliferation becomes spermatogonial (self-)renewal.
three transcriptionally distinct populations of germ This is especially true in those rodents (including
cells in the male fetus and transition from one state to the mouse and rat) where spermatogenesis proceeds
the following involves dramatic changes in tran- almost without a delay once the germ cells have
scriptome (). In rodents, the developmental dynamics settled on the basal lamina of testis cords. Spermatogenic
are somewhat different, as gonocytes first divide actively, differentiation of gonocytes/early spermatogonia
then enter a phase of mitotic quiescence, and re- can, however, be unambiguously determined by the
sumption into the cell cycle coincides with differenti- onset of stimulated by retinoic acid gene  (Stra)
ation into spermatogonia. Gonocytes-to-spermatogonia and Kit expression ().
transition involves remarkable changes in cell shape, Mouse gonocytes remain proliferatively active in

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


size, gene/protein expression, and intratubular location. the nascent testis and their numbers increase expo-
In humans, fetal and prepubertal differentiation of the nentially at least until E. (M-gonocytes) ().
germline is characterized by asynchronicity, as gonads However, markers that are associated with cell cycle
of both sexes simultaneously contain at least two de- arrest are enriched at E., and gonocytes enter
velopmentally distinct subpopulations of germ cells mitotic quiescence by E. to E. (T-gonocytes) in
(). In this section, we first concentrate on rodent fetal an asynchronous (and strain-dependent) fashion and
and early postnatal germ cell development because it do not resume cell cycle before birth (, , ,
helps to understand the dynamics and the molecular ). Mitotic reactivation (T-gonocytes) takes place
regulation of the same events in humans. In the in a strain-dependent manner ~ day after birth and
latter part, human prespermatogenic germ cells are in according to recent data concurs with gonocytes-to-
focus. spermatogonia transition (GST) (Fig. a) (, ).
GST involves dramatic changes in germ cell shape,
Rodent prespermatogenesis intracordal position, and transcriptome/translatome.
In mice, the first PGCs arrive at the presumptive gonad The gonocyte is a large, spherical cell with a
at ~E, and most of the PGCs have colonized the GRs prominent nucleus and it is located at the center of
by E. (, ). When Sry is induced, gonadal PGCs the testis cord, whereas spermatogonia are semi-
are incorporated into testis cords starting at E and circular cells at the periphery of seminiferous tubules.
from then on the germ cells that are present in the Upon GST, and concomitantly with the onset of
testis cords are referred to as gonocytes or prosper- relocation from the center to the periphery of testis
matogonia (, , ). The term prespermato- cords, perinatal germ cells start to express markers
genesis is sometimes used to denote the phase of that are associated with SSCs in the adult mouse:
testicular development involving gonocytes (, ). GDNF family receptor a (GFRa), rearranged
Furthermore, to dissect the gonocyte population into during transfection (RET), and promyelocytic leu-
functional categories, appendixes M-, T-, and T- are kemia zinc finger (PLZF, officially ZBTB) ().
occasionally used. The term M-gonocytes (M for The antimeiotic gene Nanos, which is expressed
multiplying; M-prospermatogonium) refers to mi- already by early mouse gonocytes (E.), is also
totically active early gonocytes, T-gonocytes (T for upregulated at the same stage (, ). NANOS is
transitional; T-prospermatogonia) are mitotically an RNA-binding protein that directly interacts with
quiescent but undergo genome-wide epigenetic the carbon catabolite repressor –negative on TATA
reprogramming (the topic of the following section), (CCR-NOT) deadenylase complex, resulting in deg-
and T-gonocytes (T-prospermatogonia) are the radation of specific RNAs. Recently it was suggested
proliferating and migratory immediate predecessors of that NANOS would act as a gatekeeper of GST and it
spermatogonia (Fig. a). would suppress the expression of spermatogonial
This nomenclature is somewhat confusing for at markers in a context-dependent fashion (). During
least two reasons: first, gonocytes differentiate in an human fetal testis development induction of NANOS
asynchronous manner, resulting in the coexistence of coincides specifically with entry into mitotic quiescence
two subsets of gonocytes at most time points of (). Notably, GST strictly does not take place syn-
prespermatogenesis; and second, recent research has chronously in the mouse testis, and whereas the first
elucidated that there is no clear demarcation between gonocytes gain contact with the basal lamina before
T-gonocytes and spermatogonia, as proliferatively birth or just after it, most gonocytes have translocated
quiescent gonocytes gain characteristics of sper- and transited into spermatogonia by postnatal day
matogonia (spermatogonia-specific molecular sig- (PND) . (, ).
nature, physical contact with the basal lamina of testis Undergoing GST is crucial for germ cell differ-
cords, and mitotic reactivation) in an unconcerted entiation, as the gonocytes that fail to migrate to the

882 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

periphery of seminiferous tubules are cleared by ap- human fetal testicular somatic cells, but preferentially by
optosis (). Germ cell apoptosis is a common event Leydig cells and to a lesser extent by Sertoli and peri-
during early postnatal testis development in both mice tubular cells ().
and rats, and despite becoming mitotically active, the RNA-binding protein NANOS acts cell autono-
total number of germ cells is roughly halved from mously to repress meiosis in gonocytes, and Nanos
PND . to . and PND  to , respectively (, ). deficiency results in male sterility due to loss of germ
In general, the perinatal testis development is similar cells via premature meiosis (, ). Interestingly, RA
in the mouse and rat but the events occur  to  days works as an FGF antagonist and downregulates
later in the rat. Nanos in both fetal and postnatal germ cells ().
Note that the spermatogonia of prespermatogenic CRIPTO is an obligate coreceptor of the autocrine and
testis represent a heterogeneous population of cells in self-enhancing Nodal signaling pathway that is tran-
terms of protein expression, mitotic activity, and cell siently active in mouse gonocytes (). Nodal sig-
fate (). While a proportion of nascent spermato- naling extends the period of pluripotency-related gene
gonia is set aside to form the pool of self-renewing expression in XY germ cells, and thus indirectly delays

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


SSCs, the remainder give rise to the differentiating the onset of differentiation. Additionally, ex vivo ex-
cells of the first round of spermatogenesis, a dis- periments have demonstrated that Nodal signaling
tinctive developmental program that apparently lacks is a potent inhibitor of meiotic entry (, ). In-
the differentiation-priming step (, ). To our terestingly, components of the Nodal pathway are
knowledge, it is not known what regulates the bi- dynamically expressed in human PGCs/gonocytes, too
furcation of cell fates in the early postnatal testis, but ().
RA signaling—a sign of spermatogenic commitment Exit from the cell cycle. Besides avoiding entry
and onset of male germ cell differentiation—is acti- into meiosis, early gonocytes also have to stop pro-
vated in a subset of mouse spermatogonia in a patch- liferating. TGFb signaling and Activin have been
like fashion as early as PND  (, ). It is likely shown to exert an antiproliferatory effect on PGCs,
that either (i) germ cells developmentally carry de- and they seem to work together to induce the mitotic
terminants that inherently predispose or preordain arrest of gonocytes, too (, –). Cyclin in-
them into either of the two destinies, or (ii) the so- hibitor p-mediated dephosphorylation of retino-
matic environment (primarily Sertoli and myoid blastoma protein (pRB) might be one of the “The gonocytes-to-
cells) dictates the cellular fate of nascent spermato- downstream mechanisms that TGFb signaling em- prespermatogonia transition
gonia by spatiotemporally regulating the availability ploys to impinge on gonocyte cycling (, , ). (GPT) in humans is a gradual
of differentiation-inducing (RA) or SSC-maintaining Recently, the multifunction prostanoid PGD process.”
agents. There is compelling evidence to support [reviewed by Rossitto et al. ()] has also been im-
both the suggested mechanisms, and heterogeneous plicated in the same process through promoting ex-
expression of receptors for differentiation/stemness- pression and nuclear localization of CDK inhibitor
promoting factors that are asymmetrically distrib- p (). PGD might also indirectly block meiotic
uted within the seminiferous epithelium might entry and support mitotic arrest by inducing Nanos
explain the differential propensities of nascent (, ).
spermatogonia into either of the developmental Loss of phosphatase and tensin homolog (Pten),
pathways (, ). Dmrt, and Dnd have been associated with formation
of testicular teratoma due to a failure to arrest gon-
Molecular control of rodent prespermatogenesis ocyte mitosis (, –). DND promotes mitotic
Escape from premature meiosis. Intracordal arrest by posttranscriptional mechanisms that likely
localization protects gonocytes from meiosis-inducing involve stabilization of p and p mRNAs (, ).
RA, and the PGCs that fail to get incorporated into the The role of DND in fetal germ cells is intriguing, for
testis cords will enter meiosis, undergo malignant in addition to contributing to the onset of mitotic
transformation (GCT), or perish (, , , ). There quiescence, DND has also been suggested to protect
are at least two mechanisms that prevent the premature these cells from apoptosis and malignant trans-
meiotic entry of gonocytes: first, somatic cells of the testis formation, maintain their identity, and suppress
(Sertoli, myoid, and Leydig cells) express CYP en- meiosis in collaboration with NANOS (–).
zymes that degrade the abundant mesonephros-derived Lastly, androgen action in vivo and in vitro has been
RA; and second, Sertoli cell–derived FGF upregulates noted to inhibit gonocyte proliferation, and RA
Nanos and Cripto, and thus maintains the expression of degradation might be important to arrest gonocyte
pluripotency-associated genes (Fig. b) (–). At mitosis (Fig. b) (, ). TGFb signaling and FGF
least in humans, the gonadal somatic cells in the male also promote the survival of quiescent gonocytes and,
also express lower level of enzymes needed in RA unlike M- or T-gonocytes, T-gonocytes undergo
synthesis than do the female equivalents, which probably apoptosis highly infrequently (, , ).
also affects the local concentration of bioavailable RA Downregulation of pluripotency genes.
(). Similar to the mouse, CYPB is also expressed by Among the numerous cell lineages of a developing

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 883


REVIEW

Figure 5. Developmental regulation of gonocytes. (a) Germ cell populations of the developing testis in the mouse. Insets shown (B–D)
refer to panels (b), (c), and (d). (b) M-gonocytes actively proliferate in the nascent testis cords that have not yet fully developed. They also
retain the expression of pluripotency-associated genes, such as Nanog, Oct4, and Sox2, due to activity of the Nodal/Cripto signaling. M-
gonocytes are protected from premature entry into meiosis by expression of antimeiosis gene NANOS2 and CYP26 activity in Sertoli cells
resulting in degradation of extragonadal RA. Nanos2 and Cripto are both FGF9 signaling targets. As a consequence of p21 and p27
upregulation and stabilization, the cell cycle of M-gonocytes is subsequently slowed down. TGFb signaling and PGD2 have been shown to
control this process extrinsically, whereas DND1 is cell-autonomously involved. AMH and testosterone (T) potentially also contribute. (c)
Gonocytes enter mitotic quiescence between E13.5 and E15.5 and become T1-gonocytes. This is accompanied by selective downregulation
of pluripotency-related genes as a result of DMRT1 and SOX4 action. Cell-extrinsically, PGD2 also contributes to this process. On the
contrary, Nanos2 and Dnmt3l are upregulated. At this stage testis cord formation has been completed and peritubular cells encase the
testis cords. (d) Perinatally, gonocytes start to display signs of GST. GST encompasses translocation of germ cells from the lumen to
the basement membrane of the testis cord, acquisition of a transcriptomic signature typical of spermatogonia (PLZF, GFRa1, RET), and cell
cycle resumption. Many factors have been implicated in control of GST, but the data supporting a specific role for SCF/KIT and PDGF
signaling in control of the gonocyte migration is rather solid. Gonocytes probably use actin-based (small spheres organized into filaments),
pseudopod-like protrusions to gain contact with the basement membrane. CDH1-mediated cell adhesion may also play a significant role in

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


gonocyte translocation. A subset of nascent spermatogonia is forced to enter spermatogenesis soon after GST by Sertoli cell–derived RA.
E2, estradiol. (e) Germ cell populations of the developing human testis. The developing testis houses two developmentally distinct
populations of fetal germ cells for almost 1 y. (f) Genome-wide reprogramming of the fetal germ cell epigenome. In the preimplantation
embryo the epigenome is demethylated and the lowest average CpG (5mC) methylation is observed at E3.5. The DNA is subsequently
remethylated and PGCs are specified in the posterior epiblast that exhibits relatively high global DNA methylation (~70%). Following
specification, PGC DNA is redemethylated in two stages (global and locus-specific demethylation), and at ~E13.5, 95% of CpG sequences
are in an unmethylated state. This period is accompanied by dynamic changes in chromatin modifications also. During the following few
days the gonocyte epigenome undergoes genome-wide de novo DNA methylation and acquires the paternal epigenetic signature.
PIWI–piRNA-mediated mechanisms involved in silencing of TEs are first implemented around this time. Late gonocytes and nascent
spermatogonia exhibit relatively high levels of global DNA methylation. Although DNA methyltransferases are active during
spermatogenesis, changes in DNA methylome during spermatogenesis are rather subtle. Dotted line indicates the postulated 5-
methylcytosine dynamics in humans. Spheres stand for available data at specific time points during human fetal development. gPGC,
gonadal PGC; M, M-gonocytes; T1, T1-gonocytes; T2, T2-gonocytes.

884 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

fetus, the germline is characterized by prolonged ex- puberty or during early adulthood they break free
pression of pluripotency-associated genes (). In from dormancy to form CIS, which then gives rise to a
mouse gonocytes, the Nodal/Cripto signaling pathway seminomatous or nonseminomatous testicular tumor.
is thought to maintain their expression in an FGF-
dependent manner a couple of days longer than in XX
germ cells (, ). Pluripotency genes are, however, Figure 5. (Continued)
selectively downregulated as gonocytes enter the mi-
totically quiescent phase between E. and E. (Fig,
c) (). Most notably, the expression of core net-
work genes of pluripotent state maintenance, that is,
Nanog, Sox, and Oct, are repressed. These three
genes have been implicated in embryonic germ cell
differentiation, proliferation, and survival, respectively
(, , ). Similar changes (with the exception of

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


SOX, whose expression is lacking altogether) are
observed in human fetal germ cells, as well, as they stop
proliferating (). Note that in addition to these genes,
numerous other pluripotency-associated genes are
intimately linked with male germline development
and maintenance, including spalt-like  (SALL) and
undifferentiated embryonic cell TF (UTFI), for in-
stance (, ).
Silencing of Nanog and Sox is at least partially due
to methylation of their promoters, whereas OCT is
downregulated by posttranscriptional mechanisms
(). By E., E., and E. the protein ex-
pressions of SOX, OCT, and NANOG, respectively,
become substantially reduced or undetectable in male
germ cells (, ). DMRT has been suggested to
repress the expression of Sox, Nanog, and Oct cell
autonomously, and using both direct and indirect
means (). Furthermore, PGD has been implicated
in downregulation of these genes by a DMRT-
independent mechanism (). Very recently, SOX
was shown to be needed for the proper differentiation
of gonocytes, and via an unknown mechanism Sox
deficiency resulted in prolonged expression of CRIPTO
and NANOG and inability of the gonocytes to induce
Nanos and DNA methyltransferase (Dnmt)l—two
markers for gonocyte quiescence ().
The inability of Sox-null mice to repress plurip-
otent gene expression might also originate from a
failure to downregulate the mechanisms that are re-
sponsible for their prolonged expression, such as
Nodal/Cripto or FGF signaling, as has been suggested
to explain the prolonged expression of these genes in a
conditional Emx-knockdown mouse model (,
). Notably, the inability to repress the expression of
these and other cellular pluripotency genes (such as
Dppa, Dppa, and Cdh) has been associated with
testicular tumorigenesis, and testicular GCTs are the
most frequent cause of cancer in men between  and
 years of age (, , ). Based on morpho-
logical and gene expression similarities, it has been
suggested that testicular GCTs are derived from
gonocytes that fail to undergo correct fetal differen-
tiation and are maintained within the testicular tissue
as pre–carcinoma in situ (CIS) cells (, ). In

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 885


REVIEW

Extrinsic control of GST. Given the signifi- SSCs is compromised and Rhox-deficient mice
cance of GST for formation of the SSC pool and display gradual and progressive spermatogenic decline
avoidance of a pre-CIS lesion, its molecular control is due to impaired SSC maintenance (). Notch sig-
surprisingly poorly characterized. Fortunately some naling in Sertoli cells is probably also involved in GST
conclusions can be drawn, nonetheless. Recently, the and, when excessive or improperly timed, it can
FGF signaling dependency of GST was demonstrated jeopardize gonocyte development (). Recently, it
(Fig. d) (). Culture of testicular fragments from was proposed that gonocyte-derived ligands [Delta-
the E. mouse in the presence of an FGF signaling like ligand  (DLL) and Jagged- (JAG)] might
inhibitor had an adverse effect on GST, and after a actually control the activity of Notch signaling in
-day culture germ cells failed to translocate to the testicular somatic cells, providing a piece of evidence
periphery of testis cords, remained mitotically qui- for reciprocal two-way signaling between the germline
escent, and did not display a transcriptomic signature and the soma in the developing testis (). GST or
typical of nascent spermatogonia (). Contrary to maintenance of nascent spermatogonia is also severely
earlier research, it was demonstrated that RA action is impaired in inducible Pelota (Pelo) knockout mice

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


dispensable for GST, and FGF signaling actually via a thus-far-unknown mechanism (Fig. d) ().
works upstream of the RA pathway and may thus
control spermatogonial differentiation (, ). Prespermatogenic germ cell differentiation
Moreover, WNT/b-catenin has recently been im- in humans
plicated in control of GST, as gonocytes, in which the As discussed above, the molecular basis of regulation
pathway is constitutively activated, are hyper- of gonocyte differentiation in mice and humans are
proliferative and strongly express cyclin-D, a post- remarkably similar. Molecular control responsible for
GST, spermatogonia-associated marker (, ). the phasic development of human fetal germ cells was
Interestingly, a subset of miRNAs involved in the recently characterized by Li et al. () who identi-
regulation of WNT/b-catenin and PTEN signaling fied . TFs that might play crucial roles as the
pathways is differentially expressed in gonocytes and master regulators of gene expression networks in
spermatogonia isolated from -day-old and - to human PGCs/gonocytes as they progress from mi-
-day-old mice, respectively (). This is an in- gratory to nonmotile mitotic and proliferatively qui-
teresting observation considering the emerging role escent phases. Despite obvious similarities at the
of WNT signaling in promoting undifferentiated molecular level, the dynamics of the process are
spermatogonial differentiation and proliferation, as somewhat different, as human fetal germ cells do not
well as the suggested role for PTEN in SSC main- collectively enter a phase of mitotic quiescence (similar
tenance (, , ). to ~E. to E. in mouse) but a subset (likely a
Both SCF/KIT and PDGF signaling have been constantly changing one) is proliferating throughout
implicated in gonocyte migration in rodents, and Sertoli fetal development (Fig. e) ().
cells secrete the ligands of the pathways, whereas the The first gonocytes migrate to the basal lamina of
receptors are located on gonocytes (–). PDGF(R) the testis cords at ~ wpc to give rise to germ cells that
signaling also stimulates cell cycle resumption in germ are referred to as prespermatogonia in humans (not to
cells upon and after GST, and might potentiate or prime be confused with mouse prospermatogonia, an al-
gonocytes to respond to RA (Fig. d) (, –). In ternative term for gonocytes). This coincides with the
vitro effects of a number of cytokines, hormones, and downregulation of pluripotency factors and although
growth factors on rodent PGC/gonocyte proliferation practically all germ cells of the first trimester embryo
and apoptosis have been studied in the past, but lack of express OCT, the number of OCT-expressing cells
in vivo research greatly lessens the significance of these decreases rapidly during the second trimester ().
findings (). Moreover, robust actin turnover in both The gonocytes-to-prespermatogonia transition (GPT)
gonocytes and Sertoli cells is important for proper in humans is a gradual process, and by midgestation
migration, and gonocytes probably employ actin-based most of the germ cells in the fetal testis have un-
pseudopod-like protrusions to gain contact with the dergone GPT (). There is a simultaneous increase
basal lamina. Actin dynamics are probably also required in NANOS expression suggesting a conserved anti-
for CDH-mediated cell adhesion that might play a meiotic role for this factor in both human and mouse
significant role in gonocyte repositioning (, ). A prespermatogenesis (, , ). As testis devel-
disintegrin and metalloproteinase (ADAM)/integrin/ opment proceeds from the earliest phases toward
tetraspanin complexes have also been implicated in midgestation, DDX expression is enhanced (, ).
translocation of the gonocytes and their subsequent OCT and DDX start to display a mutually exclusive
anchoring to the basal lamina (). pattern of expression, and although OCT-positive
Interestingly, most of the above-mentioned cells are heterogeneously distributed within the testis
mechanisms are dysregulated in the mutant testes cords, DDX-expressing cells are preferentially located
that lack reproductive homeobox  (Rhox). As a at the periphery (, , ). DDX thus becomes a
result, gonocyte migration and differentiation into marker for prespermatogonia, whereas OCT is

886 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

preferentially expressed by the gonocytes. Notably, total germ cell number in the male roughly doubles
% of DDX-positive cells in an -wpc fetal testis do in a week during  to  wpc, but the growth rate is
not express the proliferation marker Ki-, which subsequently reduced and it takes twice the time to
makes DDX a good marker for mitotically inactive double the number of germ cells during  to  wpc
germ cells at this stage and suggests that early pre- (). Finally,  weeks are needed to give rise to a
spermatogonia are mitotically quiescent (). In- twofold increase in germ cell count during  to
terestingly, prespermatogonia exist as syncytia of two  wpc. The number of Ki-–positive germ cells then
or more cells connected by cytoplasmic bridges, a steadily decreases toward birth, but a stage that would
characteristic that is shared with spermatogenic cells be characterized by very low germ cell mitotic activity
(, ). or total quiescence cannot be discerned. This might be
Until  wpc Ki- is predominantly expressed by masked by the asynchronous nature of the process and
the OCT-positive gonocytes. Subsequently, both coexistence of gonocytes and prespermatogonia dur-
gonocytes and prespermatogonia display a compara- ing an extended period of time. A subset of human
ble mitotic activity, but with advancing age pre- fetal germ cells at a specific developmental phase

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


spermatogonia become proliferatively inactive and the (having undergone GPT, for instance) might enter a
few germ cells that proliferate are almost without mitotically quiescent phase to undergo epigenetic
exception centrally located OCT-expressing gono- reprogramming.
cytes (). Interpretations of these data are that The number of somatic cells in the male embryo
prespermatogonia upregulate DDX and down- increases at a comparable rate to that of germ cells
regulate OCT, enter short-term mitotic quiescence, until  wpc, but then the growth slows down (). In
resume cell cycle for a short period at the end of the their data set, Li et al. () noted that somatic cells
second trimester and the beginning of the third tri- of the fetal gonads generally expressed fewer cell
mester, and then enter another proliferatively quies- cycle–related genes than did the neighboring germ
cent period before birth. OCT expression persists cells, and only ~% of somatic cells actively pro-
into early postnatal life ( to  months) and is almost liferated. Prepubertal ( to  years) development of
exclusively seen in centrally located gonocytes, making germ cells is characterized by variable mitotic activity
fetal germ cell differentiation in humans a pro- and isolated phases of spermatogonial proliferation
“Resetting of the germ cell
nouncedly asynchronized process (, ). This is in and involution that may be accompanied by emer-
epigenome during embryonic
strict contrast to rodent prespermatogenesis where gence of scarce meiotic or postmeiotic germ cells and fetal development is
OCT expression is lost in quiescent gonocytes by (–). The number of spermatogonia is slightly critical for perpetuation of the
E. and E. in mice and rats, respectively (, reduced during the first  years of life, but it is then species.”
). increased to reach a plateau by  years of age ().
As discussed above, prespermatogenic germ cell The early reduction in spermatogonial numbers might
development does not proceed in synchrony in ro- be due to apoptosis of gonocytes (mistaken as sper-
dents either. However, the scale of asynchronicity matogonia) after minipuberty, whereas the increase
between humans and mice or rats is dramatically that is seen during  to  years of age reflects the
different, and a considerably longer timespan (up to proliferation of type A spermatogonia and their dif-
 year) is required for it to conclude in humans. ferentiation into type B spermatogonia (). In total,
NANOG expression in the human fetal and early the number of germ cells is increased during infancy,
postnatal testis follows a similar pattern to that of and the testicular volume of -year-old boys is
OCT, and practically all germ cells of the early second twofold to threefold higher than in newborns (,
trimester fetus express it (). Minipuberty, charac- ). The onset of puberty, however, is characterized
terized by a surge in gonadotropin and reproductive by exponential growth of the spermatogonial pop-
hormonal activity, takes place soon after birth (at ~ to ulation and testis volume (, ).
/ months of age) and is considered the final stage of
gonocyte differentiation into infantile spermatogonia
(–). Genome-Wide Reprogramming of
the Epigenome
Prespermatogenic germ cell expansion
In humans, migrating PGCs are proliferatively active Epigenetic marks and factors collectively contribute to
but their cell cycle is accelerated once they have the molecular memory of a cell. They ensure that a cell
reached the GR and been enclosed within the testis faithfully executes the transcriptional program needed
cords as gonocytes. The number of germ cells in the for the fulfillment of its task and maintenance of its
male embryo has been suggested to increase from function. The epigenetic marks of a differentiated
~ at the beginning of the fifth embryonic week to somatic cell are relatively stable, but the epigenome
, in the -week-old embryo (). Until midg- undergoes extensive reprogramming in the mam-
estation the increase in germ cell number in the malian preimplantation embryo and the germline
human fetus follows a curvilinear pattern (). The (, ). To generate a totipotent state, epigenetic

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 887


REVIEW

marks need to be removed during early development. Soon following specification, the DNA of nascent
Gametogenesis also calls for erasure of parental epi- mPGCs is redemethylated to around half of the levels
genetic memories and removal of potential epi- that are observed in the epiblast cells. This is called
mutations from the germline (). There are also stage I (or global) DNA demethylation and it is fol-
~ imprinted genes in mammals and most of them lowed by stage II, encompassing locus-specific DNA
are found in clusters (). The proper imprinted demethylation commencing at ~E. and concluding
expression of these genes is controlled by imprinting at E. when DNA methylation during fetal germ cell
control regions. In the male germline, maternal and development reaches its lowest point (, ). At
paternal imprints are erased in PGCs and early E. ~% of cytosines at CpG sequences in genomic
gonocytes and re-established during fetal development DNA are in an unmethylated state, whereas in E.
(, ). Resetting of the germ cell epigenome epiblast CG dinucleotides carry a methyl residue in ~
during embryonic and fetal development is critical for out of  cases (, ). Consequently, almost all
perpetuation of the species. genomic features become hypomethylated. PGC
DNA methylation typically acts to repress gene epigenetic reprogramming entails not only global

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


transcription. Many genes (such as meiotic genes) DNA demethylation but also erasure of genomic
in the mammalian genome exhibit a gonad-specific imprints, reactivation of the X chromosome, and
pattern of expression and only take part in gameto- genome-wide reorganization of histone modifications
genesis. Other tissues and physiological processes must (, , –).
therefore employ mechanisms (such as DNA meth- In the process of PGC epigenetic reprogramming the
ylation) to repress the ectopic expression of these genome becomes demethylated by both replication-
genes. Failure to demethylate the DNA that hosts these independent and replication-dependent mechanisms
genes or their regulatory regions (promoters and (). The former refers to active conversion of
enhancers) in fetal germ cells might impede their -methylcytosines (mCs) to -hydroxymethylcytosines
expression during gametogenesis and result in failure (hmCs) by ten-eleven translocation methylcytosine
to produce sperm or eggs. Fetal germ cells undergo dioxygenase enzymes (TET), whereas the latter seem-
genome-wide reprogramming of the epigenome that ingly passive mechanism involves repression of DNA
erases the epigenetic memory of the previous gener- methyltransferase action: DNA methylation in pro-
ation and is postulated to create a blank slate upon liferating PGCs is not maintained owing to suppression
which sex-specific mechanisms then work to establish of enzymes that carry out maintenance or de novo DNA
an androgenetic or gynogenetic (in males and females, methylation (, –). As DNA gets replicated,
respectively) epigenetic signature. CpGs of the daughter strand are not methylated, and
Activation of germline genes in nascent gonocytes consequently every round of DNA replication halves the
is partially mediated by DNA demethylation. Despite total number of methylated CG dinucleotides per unit of
global hypomethylation of the genome, transcription DNA, a phenomenon that is also called replication-
is not promiscuously activated in PGCs (, ). coupled dilution.
PGCs must therefore recruit other mechanisms to
avoid uncontrolled transcription of DNA, including Demethylation in mPGCs
activation of transposable elements (TEs). However, In the mouse, replication-coupled dilution relies on
not everything gets demethylated, and there are ge- BLIMP and PRDM, two key genes in mPGC
nomic regions that at least partially escape the resetting specification, which repress the expression of de novo
of the epigenome, potentially contributing to trans- DNMTs Dnmta and Dnmtb, and ubiquitin-like with
generational epigenetic inheritance (TEI). PHD and ring finger domains  (Uhrf), a factor
needed to recruit DNMT, the maintainer of cellular
Global demethylation of PGC genome DNA methylation signature (, , ). This
The epigenetic marks on the zygotic DNA need to be mechanism is likely to be activated in mPGCs upon
extensively erased to reset the epigenome of cells of the specification (stage I), whereas mC-to-hmC con-
early embryos for naive pluripotency (Fig. f). In the version that accounts for erasing imprints and
preimplantation embryo the epigenome undergoes demethylation of meiotic gene promoters does not
early demethylation and the lowest average CpG become significant before mPGCs settle in the gonadal
methylation of all preimplantation embryonic stages is primordia (stage II) (, , , ). DNA
observed in the inner cell mass of an E. mouse methylation at imprinting control regions and meiotic
blastocyst (). This is, however, a transient state and gene promoters is preserved by DNMT, an action
the DNA is subsequently and rapidly remethylated. that is needed to prevent precocious differentiation of
mPGCs are then derived from the proximal epiblast of gonocytes ().
the early postimplantation embryo and they exhibit Three TET enzymes (TET to TET) have been
global DNA methylation levels similar to their somatic implicated in hydroxylation of mC, but TET and
neighbors that are primed for lineage differentiation TET have been considered to play the primary role in
(, , ). the process, whereas TET is probably dispensable for

888 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

mC-to-hmC conversion in the germline (, ). in chromatin modifications during the course of male
Upregulation of TET and TET in mPGCs at E. is germ cell epigenetic reprogramming ().
followed by an increase of hmC levels that reach their The levels of a repressive histone mark trimethy-
peak at ~E (, ). Cells can replace hmC (or its lation of lysine  on histone  (HKme) persis-
derivatives) enzymatically by unmethylated cytosines, tently increase in migratory mPGCs, are maintained in
but data suggest that the decline of hmC after E. is nascent gonocytes, and are downregulated as gono-
mainly due to replication-coupled dilution (, , cytes enter mitotic quiescence (Fig. f) (, , ).
). The significance of TET-mediated demethyla- Concomitantly with enhancement of HKme,
tion was recently severely questioned and it was there is a loss of dimethylation of lysine  on histone
suggested that the canonical demethylating activity of H (HKme; a repressive mark) in mPGCs (,
TET is restricted to removal of aberrant residual ). Whereas HKme behaves similarly in hPGCs,
DNA methylation and protection of newly deme- HKme is only transiently enriched in migratory
thylated DNA from reacquiring this mark. Moreover, germ cells and depleted in hPGCs upon GR coloni-
TET was implicated in a crucial role in germline zation. The significance of this difference at the mo-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


development through activation of germline genes in lecular level is not understood. Dimethylations of
gonocytes via a DNA demethylation-independent arginine  on HA and H (HA/HRme) are also
mechanism (). increased in migrating mPGCs but downregulated
upon arrival at the embryonic gonad (Fig. f) ().
Demethylation in hPGCs Notably, HA/HRme levels do not fluctuate
hPGCs follow essentially similar dynamics in terms of during hPGC development (). HA/HRmes
DNA demethylation as described above for mPGCs, have been suggested to be critical for the repression
but some differences do exist and not all observations of TEs in early PGCs that have lost HKme and
in mice can be faithfully extrapolated to humans (, do not yet have an active Piwi-interacting RNA
, ). First, although repression of DNMTA, (piRNA)–mediated mechanism for TE silencing
DNMTB, and UHRF are associated with replication- (discussed below) (). Notably, the PIWI–piRNA
coupled loss of CG dinucleotide methylation in pathway might be active in hPGCs relatively early, as
hPGCs, too, partially or completely different factors the key components of the pathway are upregulated in
must be at play, as PRDM is likely indispensable for - to -wpc hPGCs and the expression is maintained “Epigenetic ground state is
the process (, , , ). Second, imprinted in both gonocytes and prespermatogonia (, ). achieved in germ cells isolated
genes undergo TET-mediated hydroxymethylation Despite nearly complete erasure of cytosine from 11-week-old male
already in migratory hPGCs, which is in line with the methylation in mPGCs and hPGCs, some DNA se- fetuses.”
data showing TET and TET induction already in quences are protected and they retain relatively high
early hPGCLCs (, , ). Thus DNA demethy- levels of DNA methylation (, , , ). Es-
lation in hPGCs cannot be subdivided into equally pecially, evolutionarily young and potentially haz-
distinct stages as has been previously considered to ardous TEs, pericentric heterochromatin, some loci
exist in mice. At around the time of GR colonization, associated with TEI, but also some single copy genes
hPGCs of the sixth embryonic week already exhibit remain methylated. DNA methylation at these escapee
substantial demethylation, and global methylation of loci might be maintained by HKme- or tripartite
CG dinucleotides is at %. Epigenetic ground state is motif containing  (TRIM)–mediated and UHRF-
achieved in germ cells isolated from -week-old male independent recruitment of DNMT (). DNA
fetuses (). At this point .% of CpGs in the methylation-mediated TEI through the germline has
genomic DNA are found unmethylated (). been under heated debate recently. However, these
marks would have to tolerate two waves of epigenetic
Other aspects of PGC epigenome reprogramming reprogramming in every generation and be susceptible
In PGCs that undergo epigenetic reprogramming, to environmental agents to qualify as carriers of trans-
DNA methylation and transcription need to be generational nongenetic information to subsequent
uncoupled, and therefore a mechanism independent generations with phenotypic consequences, and
of DNA methylation is required to protect PGCs from therefore the significance of DNA methylation in TEI
the harmful effects of aberrant gene expression and has justifiably been questioned ().
activation of TEs [reviewed by Tang et al. ()]. This
task has been assigned to chromatin modifications, Acquisition of paternal epigenetic signature
and accordingly, there is a dynamic turnover of histone Knowledge regarding the mechanisms of remethyla-
marks in PGCs at and around the time of low global tion during gonocyte/fetal germ cell quiescence is still
mC levels (Fig. f). Species-to-species variation in the in its infancy. This is especially true for human fetal
exact means that are used, however, is typical. In- germ cells. De novo DNA methylation is a genome-
terestingly, the factors (BLIMP/PRDM) whose wide process and it also involves the three known
action is needed to lower DNA methylation in a paternally methylated germline differentially methyl-
genome-wide fashion also drive the dynamic changes ated regions, but not much is known about how

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 889


REVIEW

paternal-specific DNA methylation imprints are mech- suggesting that the methylation pattern of germ cells is
anistically established in the male germline. largely established prior to onset of spermatogenesis
In the male mouse germline, prenatal de novo ().
methylation of DNA starts at ~E. and is completed As discussed earlier in this text, fetal germ cell entry
by E. (, , ). Although there is considerable into mitotic quiescence is accompanied with dramatic
variation, by E. gonocytes display on average % changes in transcription in both humans and mice. In
methylation of CG dinucleotides. During subsequent the mouse, DNMTs (Dnmta, Dnmtb and Dnmtl) are
differentiation the DNA of male germ cells gets further among the genes whose expression is strongly induced in
methylated and spermatozoa display % to % gonocytes between E. and E. (, , ). This
methylation at CpG sites (, ). Interestingly, DNA coincides with methylation of Nanog and Sox pro-
methylome does not markedly change between moters that is associated with their transcriptional re-
adult human spermatogonial stem cells and sperm, pression in the male germline (). These data suggest
that repression of pluripotency-associated genes is due to
global remethylation of the gonocyte genome. In-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


terestingly, DNMTA, but not DNMTB, has been
shown to be essential for de novo DNA methylation of
the imprinted loci in germ cells of both sexes (). In the
human fetal testis, the highest levels of DNMTA and
DNMT are recorded during the nd developmental
week but the levels are relatively high throughout weeks
 to  (). Owing to the paucity of knowledge we can
only presume that this is the period when human
prespermatogonial DNA is remethylated and the pa-
ternal imprints are established. Low methylation
levels are maintained in male PGCs at least until 
wpc (). Although the germ cell DNA is extensively
remethylated already during prenatal development,
the spermatogenic cell genome also undergoes dy-
namic changes in methylation in mitotic and early
meiotic cells (). Interestingly, spermatogenic
differentiation commitment within the mouse
germline is associated with strong upregulation of
DNMTA and DNMTB (). Their increased
expression probably contributes to the irreversibility
of the process, as differentiating spermatogonia
cannot revert back to the self-renewing state.
Restoration of DNA methylation is important not
only to establish the paternal epigenetic signature but
also to reinstate the control on gene expression. Gon-
ocytes and their developmental successors also employ
other means to repress transcription. A male germline-
specific mechanism of TE silencing is recruited in the
mouse embryo at E. when the expression of MILI, a
PIWI family protein, is first observed in gonocytes (Fig.
f) (). MILI interacts with piRNAs to carry out its role
in transposon mRNA and gene silencing (, ).
Another PIWI family member, MIWI, follows a couple
of days later. Although MILI expression is maintained in
Figure 6. The number of Sertoli cells during childhood and puberty in primates based on studies
by Rey et al., 1993 (500), Simorangkir et al., 2012 (504), and Cortes et al., 1987 (499). The number of the male germline until postmeiotic cells, MIWI is
adult subjects (all .25 years of age) are shown at age 18 in the lowest panel. The data were transiently expressed and is depleted from most male
extracted from original publications using WebPlot Digitizer v4.18. [Data from Cortes D, Muller J, mouse germ cells by  days after birth and displays a
Skakkebaek NE. Proliferation of sertoli cells during development of the human testis assessed by restricted pattern of expression within the differentiation-
stereological methods. Int J Androl 1987;10(4):589–596; from Rey RA, Campo SM, Bedecarras P, et al. primed subset of undifferentiated spermatogonia (,
Is infancy a quiescent period of testicular development? Histological, morphometric, and functional
). Notably, the effect of piRNAs on TE silencing
study of the seminiferous tubules of the cebus monkey from birth to the end of puberty. J Clin
Endocrinol Metab 1993;76(5):1325–1331; and from Simorangkir DR, Ramaswamy S, Marshall GR,
is through de novo DNA methylation, and the
et al. Sertoli cell differentiation in rhesus monkey (macaca mulatta) is an early event in puberty and PIWI–piRNA pathway is therefore tethered into
precedes attainment of the adult complement of undifferentiated spermatogonia. Reproduction transcriptional control via DNA methyltransferase
2012;143(4):513–522.] action ().

890 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Postnatal Testicular Growth cells before puberty (). Thus, before the onset of
puberty testicular volume provides an estimate of the
Sertoli cell proliferation and maturation Sertoli cell numbers, assuming that the interindividual
In adulthood, the number of Sertoli cells correlates differences in interstitial volume are small. Previously,
both with testicular volume and sperm output both in testicular volume has been measured in autopsy studies
rodents and in humans (, ). Sertoli cells provide by weighing or by water displacement (, , ).
the physical framework for adult testicular micro- In more recent studies ultrasound has been used
architecture, habitats, and regulation of germ cells, and (–), which allows longitudinal follow-up.
their intercellular junctions facilitate and form the bulk Inhibin B. Inhibins are testicular peptides that
of the blood–testis barrier. First, the overall events that are generally thought to inhibit the activity of the
take place in Sertoli cell proliferation and maturation as hypothalamus (). Inhibin B is the predominant
well as the circulating markers of Sertoli cell function are inhibin in primates and it is expressed predominantly
briefly described. Then, the changes during the three in the Sertoli cells before and as a joint product of
developmental periods, that is, the neonatal period, Sertoli cells and spermatocytes after the onset of

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


juvenile period, and puberty, are reviewed in more detail. spermatogenesis (, ).
The circulating inhibin B concentration correlates
Pattern of Sertoli cell proliferation in with the Sertoli cell number in rodents, and the
autopsy studies
Counting of Sertoli cells in histological sections under
stereomicroscopy is the gold standard for their quan-
tification. However, such data are scarce as, in humans,
they require autopsy studies. A cross-sectional autopsy
study suggests that the increase in Sertoli cell number
seems to take place mostly during the first neonatal
months (i.e., minipuberty) and during puberty ().
However, the results of this study are limited by the
small number of subjects, large interindividual differ-
ence in the number of Sertoli cells, and the inevitable
cross-sectional nature of the study.
The importance of puberty and minipuberty in
Sertoli cell proliferation is supported by experimental
studies in nonhuman primates. In Cebus monkeys and
marmosets, the number of Sertoli cells increases sharply
during minipuberty, and a slight increase was found in
early puberty (, ). In contrast, the increase in
Sertoli cell number in rhesus monkeys during puberty is
rapid, whereas a little increase in their number takes
place during the neonatal period (–). The
numbers of Sertoli cells during primate lifetimes based
on autopsy and experimental studies are shown in Fig.
. Thus, the relative importance between either of the
two periods for the final Sertoli cell number in humans
remains uncertain, especially as the experimental in-
hibition of Sertoli cell proliferation during the mini-
pubertal period in marmosets does not seem to translate
to reduced Sertoli cell number in adulthood ().

Biomarkers of Sertoli cell number and maturation


Substantial interindividual variability has been observed
in the number of Sertoli cells in adulthood. Less invasive
biomarkers of Sertoli cells provide a good picture on
Sertoli cell development, as they allow longitudinal
follow-up. The longitudinal pattern of testicular growth
by ultrasonography, circulating inhibin B, and AMH as
well as their temporal relationship with Sertoli cell
number in humans are shown in Fig. .
Testicular volume. In Cebus monkeys, ~% of Figure 7. The longitudinal pattern of biomarkers of Sertoli cell proliferation and maturation in
the testicular cells in the seminiferous tubules are Sertoli humans and their temporal relationship with the number of Sertoli cells.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 891


REVIEW

hemicastration halves the circulating concentration in Sertoli cell proliferation and maturation
rhesus monkeys () and humans (). In humans, during minipuberty
the inhibin B concentration correlates with testicular During the first postnatal months, there is an ap-
volume during minipuberty (). proximately fourfold increase in the number of Sertoli
AMH. AMH is an interesting circulating cells in humans and rhesus monkeys, although the
marker in terms of functional maturation. The ex- increase is even greater in Cebus monkeys (, ).
pression of AMH is switched on early during the fetal This growth is caused by a similar increase in the length
Sertoli cell development, and it causes regression of the of seminiferous tubules, whereas the diameter of the
Müllerian ducts (). During the fetal, neonatal, and seminiferous tubules remains unaltered (, , ).
juvenile periods, AMH is secreted exclusively by the The increase in the number of Sertoli cells during
Sertoli cells (). The levels of AMH peak during minipuberty seems to be driven both by proliferation
minipuberty, and later very early during puberty at  to and protection from apoptosis, and it seems to take
 years (, ), consistent with the observed place mostly during the first postnatal month in
testicular growth and the increase in Sertoli cell humans (). This results in a marked increase in the

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


numbers. The loss of AMH secretion occurs early size of the testes. Based on autopsy studies (shown in
during puberty (, ), as a result of the increase in Fig. ) and studies utilizing ultrasonography to assess
intratesticular testosterone concentration and the testicular volume, testicular growth takes place until
entry of germ cells into meiosis already at the age when the age of  months (–). A cross-sectional
circulating testosterone concentrations are low and/or study utilizing ultrasonography reported that tes-
indistinguishable (, ). ticular volume may even decrease after the age of
GnRH, FSH, and LH. Hypothalamic GnRH  months ().
and gonadotropic FSH and LH are not direct biomarkers There are several candidates for factors that drive
of Sertoli cell function. However, the peak in activity of Sertoli cell development during minipuberty. The
the HPG axis during minipuberty and puberty clearly obvious first candidates are the gonadotropins FSH
coincides with the periods of Sertoli cell proliferation. and LH. Experimental silencing of the HPG axis by
Furthermore, a compensatory increase in FSH levels administration of a GnRH antagonist is associated
often indicates a Sertoli cell dysfunction or clearly low with a % reduction in Sertoli cell number by the end
numbers of Sertoli cells. During the juvenile presumed of minipuberty (). Furthermore, FSH/LH supple-
hiatus between minipuberty and puberty, the nocturnal mentation of baby boys with hypogonadotropic
peaks in GnRH in circulation seem to be of importance hypogonadism markedly increases the testicular vol-
for the functional maturation of Sertoli cells. ume and circulating concentrations of inhibin B and
AMH according to several case reports (–).
The FSH receptor is expressed already starting from
the fetal period (). Men with an inactivating FSH
receptor mutation have reduced semen quality and
testicular volume in adulthood, compatible with the
diminished minipubertal proliferation of Sertoli cells
(), although this effect may also be caused later
during testicular development. It remains still unknown
whether the LH/androgen secretion contributes to
the proliferation of Sertoli cells during minipuberty.
Circulating AMH levels are higher during mini-
puberty among subjects with androgen insensitivity,
suggesting that the androgens might affect the ex-
pression of AMH. Neonatal Sertoli cells do not
express AR before the onset of puberty in humans
and nonhuman primates (, , ). Thus, the
potential effect of LH/androgens has to be relayed
either by Leydig cells or PMCs, which do express
the AR.

Figure 8. Testicular growth during minipuberty based on autopsy studies by Berensztein et al., Sertoli cell proliferation and maturation during
2002 (506) and Bidlingmaier et al., 1983 (505). The data were extracted from original publications the juvenile period
using WebPlot Digitizer v4.18. [Data from Bidlingmaier F, Dorr HG, Eisenmenger W, et al.
Although the period between minipuberty and true
Testosterone and androstenedione concentrations in human testis and epididymis during the first
two years of life. J Clin Endocrinol Metab 1983;57(2):311–315; and from Berensztein EB, Sciara MI,
puberty is characterized by silence of the HPG axis
Rivarola MA, Belgorosky A. Apoptosis and proliferation of human testicular somatic and germ cells and, for example, lack of circulating testosterone, it has
during prepuberty: High rate of testicular growth in newborns mediated by decreased apoptosis. J been suggested that during this period the testis might
Clin Endocrinol Metab 2002;87(11):5113–5118.] remain “silently active” (). Especially toward the

892 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

end of the juvenile period, the GnRH axis starts to Interestingly, when comparing the studies, the peak
exhibit pulsatile activity nocturnally. and loss of AMH appears to begin already before the
During this hiatus in activity of the HPG axis, testicular volume of $ mL by orchidometer, which is
testosterone is undetectable and inhibin B and AMH often considered a marker of pubertal onset, as it is the
levels first decline from the peak levels during mini- first reliable marker of the imminent peak in testicular
puberty, and then remain stable (, ). A small growth. Our longitudinal study suggests that clear
proportion of Sertoli cells seems to proliferate in testicular growth can be shown by ultrasonography long
rhesus monkeys during the juvenile period between before the attainment of testicular volume $ mL.
minipuberty and true puberty (). Owing to the Thus, such testicular growth might be a marker of
relatively small number of subjects in autopsy studies, Sertoli cell proliferation (). Studies in XXSxrb mice
it remains unknown whether a small increase in Sertoli (a mouse line displaying premeiotic failure in sper-
cell number takes place in humans as well (). matogenesis due to presence of two X chromosomes)
However, relative to minipuberty or true puberty, the suggest that the downregulation in AMH seems to be
contribution of the juvenile period does not appear complemented by the entry into meiosis, as AMH levels

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


that significant. Alternatively, this period seems im- remained elevated despite the typical pubertal mor-
portant for the maturation of Sertoli cells, which in- phological changes ().
cludes changes in morphology and expression of Adulthood. Until recently, there has been a
various enzymes. general agreement that Sertoli cells cease to proliferate
Puberty. This period coincides with the reac- during puberty, and thus they might not proliferate
tivation of the hypothalamic–pituitary axis, which during adulthood. A recent study suggested that
initially occurs nocturnally. The small number of Sertoli cells of the seminiferous tubules in the tran-
subjects in studies precludes careful analysis of when sitional zone near to rete testis may elongate also in
the peak in Sertoli cell proliferation occurs. However, adulthood in rats (). However, it is not known if
Sertoli cell proliferation takes place early during the this is true in humans. In young adults, sperm pro-
pubertal transition in rhesus monkeys, when the duction does not increase between  and  years of
circulating nocturnal testosterone concentrations had age, which reflects the cessation of Sertoli cell pro-
not exhibited a robust pubertal increase (). liferation at puberty and implicates that sperm pro-
Among men with hypogonadotropic hypogonadism, duction cannot be augmented in adult men (). An
the gonadotropin substitution with FSH alone in- exception to this rule is hypogonadotropic hypo-
creases the testicular volume and the circulating gonadism that prevents puberty, and gonadotropin
levels of inhibin B and AMH (–). FSH in- treatment is needed for the initiation of puberty and
creases the proliferation of Sertoli cells in humans, induction of spermatogenesis in adulthood.
although both LH and FSH are able to stimulate the
proliferation of Sertoli cells in late juvenile rhesus
monkeys (). Concluding Remarks
After the possible peak or plateau in AMH, the
levels decline rapidly while a concomitant increase in Rapid development of research technologies has ad-
inhibin B levels takes place (, , ). The levels vanced our knowledge on testis development and
of AMH inversely correlate with circulating an- function. Molecular mechanisms of DSDs are now
drogen levels and remain persistently elevated in known for a large number of syndromes. Emerging
mice or boys who lack a functional AR (, ). new data on epigenetics will be important for further
Additionally, the treatment of men with hypo- understanding of disease mechanisms. Such data may
gonadotropic hypogonadism with the combination also explain complex interactions between genes and
of FSH and LH/human chorionic gonadotropin de- the environment. Comprehensive research on genes,
creases AMH levels (). In contrast, treatment with epigenetic mechanisms, and environmental factors,
FSH alone increases the circulating levels of AMH in including lifestyle and endocrine disruptors, is needed
men with hypogonadotropic hypogonadism (). to solve many open questions.

References
1. Extavour CG, Akam M. Mechanisms of germ cell 3. Lawson KA, Dunn NR, Roelen BA, Zeinstra LM, primordial germ cells in the mouse. Mol Endocrinol.
specification across the metazoans: epigenesis and Davis AM, Wright CV, Korving JP, Hogan BL. Bmp4 is 2000;14(7):1053–1063.
preformation. Development. 2003;130(24):5869–5884. required for the generation of primordial germ cells 5. Ying Y, Zhao GQ. Cooperation of endoderm-
2. Witschi E. Migration of germ cells of human in the mouse embryo. Genes Dev. 1999;13(4): derived BMP2 and extraembryonic ectoderm-
embryos from the yolk sac to the primitive go- 424–436. derived BMP4 in primordial germ cell
nadal folds. Contr Embryol Carnegie Inst. 1948;209: 4. Ying Y, Liu XM, Marble A, Lawson KA, Zhao GQ. generation in the mouse. Dev Biol. 2001;232(2):
67–80. Requirement of Bmp8b for the generation of 484–492.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 893


REVIEW

6. Chang H, Matzuk MM. Smad5 is required for specification in mice. Nat Cell Biol. 2013;15(8): 38. Fujiwara Y, Komiya T, Kawabata H, Sato M,
mouse primordial germ cell development. Mech 905–915. Fujimoto H, Furusawa M, Noce T. Isolation of a
Dev. 2001;104(1–2):61–67. 22. Saitou M, Barton SC, Surani MA. A molecular DEAD-family protein gene that encodes a murine
7. Hayashi K, Kobayashi T, Umino T, Goitsuka R, programme for the specification of germ cell fate in homolog of Drosophila vasa and its specific ex-
Matsui Y, Kitamura D. SMAD1 signaling is critical mice. Nature. 2002;418(6895):293–300. pression in germ cell lineage. Proc Natl Acad Sci USA.
for initial commitment of germ cell lineage from 23. Buehr M, McLaren A, Bartley A, Darling S. Pro- 1994;91(25):12258–12262.
mouse epiblast. Mech Dev. 2002;118(1–2):99–109. liferation and migration of primordial germ cells in 39. Lim AK, Lorthongpanich C, Chew TG, Tan CW,
8. Chu GC, Dunn NR, Anderson DC, Oxburgh L, We/We mouse embryos. Dev Dyn. 1993;198(3): Shue YT, Balu S, Gounko N, Kuramochi-Miyagawa S,
Robertson EJ. Differential requirements for Smad4 182–189. Matzuk MM, Chuma S, Messerschmidt DM, Solter
in TGFb-dependent patterning of the early mouse 24. Tsuda M, Sasaoka Y, Kiso M, Abe K, Haraguchi S, D, Knowles BB. The nuage mediates retro-
embryo. Development. 2004;131(15):3501–3512. Kobayashi S, Saga Y. Conserved role of nanos transposon silencing in mouse primordial ovarian
9. Arnold SJ, Maretto S, Islam A, Bikoff EK, Robertson proteins in germ cell development. Science. 2003; follicles. Development. 2013;140(18):3819–3825.
EJ. Dose-dependent Smad1, Smad5 and Smad8 301(5637):1239–1241. 40. Hill PWS, Leitch HG, Requena CE, Sun Z, Amouroux
signaling in the early mouse embryo. Dev Biol. 2006; 25. Youngren KK, Coveney D, Peng X, Bhattacharya C, R, Roman-Trufero M, Borkowska M, Terragni J,
296(1):104–118. Schmidt LS, Nickerson ML, Lamb BT, Deng JM, Vaisvila R, Linnett S, Bagci H, Dharmalingham G,
10. Ohinata Y, Payer B, O’Carroll D, Ancelin K, Ono Y, Behringer RR, Capel B, Rubin EM, Nadeau JH, Matin Haberle V, Lenhard B, Zheng Y, Pradhan S, Hajkova
Sano M, Barton SC, Obukhanych T, Nussenzweig M, A. The Ter mutation in the dead end gene causes P. Epigenetic reprogramming enables the transition

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


Tarakhovsky A, Saitou M, Surani MA. Blimp1 is a germ cell loss and testicular germ cell tumours. from primordial germ cell to gonocyte. Nature.
critical determinant of the germ cell lineage in mice. Nature. 2005;435(7040):360–364. 2018;555(7696):392–396.
Nature. 2005;436(7048):207–213. 26. Chiquoine AD. The identification, origin, and mi- 41. Hoffman JA, Wu CI, Merrill BJ. Tcf7l1 prepares
11. Vincent SD, Dunn NR, Sciammas R, Shapiro-Shalef gration of the primordial germ cells in the mouse epiblast cells in the gastrulating mouse embryo for
M, Davis MM, Calame K, Bikoff EK, Robertson EJ. embryo. Anat Rec. 1954;118(2):135–146. lineage specification. Development. 2013;140(8):
The zinc finger transcriptional repressor Blimp1/ 27. Ginsburg M, Snow MH, McLaren A. Primordial 1665–1675.
Prdm1 is dispensable for early axis formation but is germ cells in the mouse embryo during gastrula- 42. Sun LT, Yamaguchi S, Hirano K, Ichisaka T, Kuroda
required for specification of primordial germ cells in tion. Development. 1990;110(2):521–528. T, Tada T. Nanog co-regulated by Nodal/Smad2
the mouse. Development. 2005;132(6):1315–1325. 28. Sato M, Kimura T, Kurokawa K, Fujita Y, Abe K, and Oct4 is required for pluripotency in developing
12. Ohinata Y, Ohta H, Shigeta M, Yamanaka K, Masuhara M, Yasunaga T, Ryo A, Yamamoto M, mouse epiblast. Dev Biol. 2014;392(2):182–192.
Wakayama T, Saitou M. A signaling principle for the Nakano T. Identification of PGC7, a new gene 43. Campolo F, Gori M, Favaro R, Nicolis S, Pellegrini M,
specification of the germ cell lineage in mice. Cell. expressed specifically in preimplantation embryos Botti F, Rossi P, Jannini EA, Dolci S. Essential role of
2009;137(3):571–584. and germ cells. Mech Dev. 2002;113(1):91–94. Sox2 for the establishment and maintenance of the
13. Yamaji M, Seki Y, Kurimoto K, Yabuta Y, Yuasa M, 29. Yabuta Y, Kurimoto K, Ohinata Y, Seki Y, Saitou M. germ cell line. Stem Cells. 2013;31(7):1408–1421.
Gene expression dynamics during germline speci- 44. Chambers I, Silva J, Colby D, Nichols J, Nijmeijer B,
Shigeta M, Yamanaka K, Ohinata Y, Saitou M. Critical
Robertson M, Vrana J, Jones K, Grotewold L, Smith
function of Prdm14 for the establishment of the germ fication in mice identified by quantitative single-cell
A. Nanog safeguards pluripotency and mediates
cell lineage in mice. Nat Genet. 2008;40(8):1016–1022. gene expression profiling. Biol Reprod. 2006;75(5):
germline development. Nature. 2007;450(7173):
14. Kurimoto K, Yabuta Y, Ohinata Y, Shigeta M, 705–716.
1230–1234.
Yamanaka K, Saitou M. Complex genome-wide 30. Yamaguchi S, Kimura H, Tada M, Nakatsuji N, Tada
45. Yamaguchi S, Kurimoto K, Yabuta Y, Sasaki H,
transcription dynamics orchestrated by Blimp1 T. Nanog expression in mouse germ cell develop-
Nakatsuji N, Saitou M, Tada T. Conditional
for the specification of the germ cell lineage in mice. ment. Gene Expr Patterns. 2005;5(5):639–646.
knockdown of Nanog induces apoptotic cell death
Genes Dev. 2008;22(12):1617–1635. 31. Weber S, Eckert D, Nettersheim D, Gillis AJ, Schäfer
in mouse migrating primordial germ cells. Devel-
15. Aramaki S, Hayashi K, Kurimoto K, Ohta H, Yabuta S, Kuckenberg P, Ehlermann J, Werling U, Biermann
opment. 2009;136(23):4011–4020.
Y, Iwanari H, Mochizuki Y, Hamakubo T, Kato Y, K, Looijenga LH, Schorle H. Critical function of AP-2
46. Zhang M, Leitch HG, Tang WWC, Festuccia N, Hall-
Shirahige K, Saitou M. A mesodermal factor, T, gamma/TCFAP2C in mouse embryonic germ cell
Ponsele E, Nichols J, Surani MA, Smith A, Chambers
specifies mouse germ cell fate by directly activat- maintenance. Biol Reprod. 2010;82(1):214–223.
I. Esrrb complementation rescues development of
ing germline determinants. Dev Cell. 2013;27(5): 32. Nakaki F, Hayashi K, Ohta H, Kurimoto K, Yabuta Y,
Nanog-null germ cells. Cell Reports. 2018;22(2):
516–529. Saitou M. Induction of mouse germ-cell fate 332–339.
16. Yu J, Angelin-Duclos C, Greenwood J, Liao J, Calame by transcription factors in vitro. Nature. 2013; 47. Murakami K, Günesdogan U, Zylicz JJ, Tang WWC,
K. Transcriptional repression by blimp-1 (PRDI-BF1) 501(7466):222–226. Sengupta R, Kobayashi T, Kim S, Butler R, Dietmann
involves recruitment of histone deacetylase. Mol 33. Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou S, Surani MA. NANOG alone induces germ cells in
Cell Biol. 2000;20(7):2592–2603. M. Reconstitution of the mouse germ cell speci- primed epiblast in vitro by activation of enhancers.
17. Gy}ory I, Wu J, Fejér G, Seto E, Wright KL. PRDI-BF1 fication pathway in culture by pluripotent stem Nature. 2016;529(7586):403–407.
recruits the histone H3 methyltransferase G9a in cells. Cell. 2011;146(4):519–532. 48. Hopf C, Viebahn C, Püschel B. BMP signals and the
transcriptional silencing. Nat Immunol. 2004;5(3): 34. Ohta H, Kurimoto K, Okamoto I, Nakamura T, transcriptional repressor BLIMP1 during germline
299–308. Yabuta Y, Miyauchi H, Yamamoto T, Okuno Y, segregation in the mammalian embryo. Dev Genes
18. Ancelin K, Lange UC, Hajkova P, Schneider R, Hagiwara M, Shirane K, Sasaki H, Saitou M. In vitro Evol. 2011;221(4):209–223.
Bannister AJ, Kouzarides T, Surani MA. Blimp1 as- expansion of mouse primordial germ cell-like cells 49. Sasaki K, Nakamura T, Okamoto I, Yabuta Y, Iwatani
sociates with Prmt5 and directs histone arginine recapitulates an epigenetic blank slate. EMBO J. C, Tsuchiya H, Seita Y, Nakamura S, Shiraki N,
methylation in mouse germ cells. Nat Cell Biol. 2006; 2017;36(13):1888–1907. Takakuwa T, Yamamoto T, Saitou M. The germ cell
8(6):623–630. 35. Kurimoto K, Yabuta Y, Hayashi K, Ohta H, Kiyonari fate of cynomolgus monkeys is specified in the
19. Su ST, Ying HY, Chiu YK, Lin FR, Chen MY, Lin KI. H, Mitani T, Moritoki Y, Kohri K, Kimura H, nascent amnion. Dev Cell. 2016;39(2):169–185.
Involvement of histone demethylase LSD1 in Yamamoto T, Katou Y, Shirahige K, Saitou M. 50. De Felici M. Origin, migration, and proliferation of
Blimp-1-mediated gene repression during plasma Quantitative dynamics of chromatin remodeling human primordial germ cells. In: Coticchio G,
cell differentiation. Mol Cell Biol. 2009;29(6): during germ cell specification from mouse em- Albertini DF, De Santis L, eds. Oogenesis. London,
1421–1431. bryonic stem cells. Cell Stem Cell. 2015;16(5): UK: Springer-Verlag; 2013:19–37.
20. Cretney E, Xin A, Shi W, Minnich M, Masson F, 517–532. 51. McKay DG, Hertig AT, Adams EC, Danziger S.
Miasari M, Belz GT, Smyth GK, Busslinger M, Nutt 36. Cooke HJ, Lee M, Kerr S, Ruggiu M. A murine Histochemical observations on the germ cells of
SL, Kallies A. The transcription factors Blimp-1 and homologue of the human DAZ gene is autosomal human embryos. Anat Rec. 1953;117(2):201–219.
IRF4 jointly control the differentiation and function and expressed only in male and female gonads. 52. Kerr CL, Hill CM, Blumenthal PD, Gearhart JD.
of effector regulatory T cells. Nat Immunol. 2011; Hum Mol Genet. 1996;5(4):513–516. Expression of pluripotent stem cell markers in the
12(4):304–311. 37. Gill ME, Hu YC, Lin Y, Page DC. Licensing of ga- human fetal ovary. Hum Reprod. 2008;23(3):
21. Magnúsdóttir E, Dietmann S, Murakami K, metogenesis, dependent on RNA binding protein 589–599.
Günesdogan U, Tang F, Bao S, Diamanti E, Lao K, DAZL, as a gateway to sexual differentiation of fetal 53. Høyer PE, Byskov AG, Møllgård K. Stem cell factor
Gottgens B, Azim Surani M. A tripartite transcription germ cells. Proc Natl Acad Sci USA. 2011;108(18): and c-Kit in human primordial germ cells and fetal
factor network regulates primordial germ cell 7443–7448. ovaries. Mol Cell Endocrinol. 2005;234(1–2):1–10.

894 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

54. Mamsen LS, Brøchner CB, Byskov AG, Møllgard K. 68. Fang F, Angulo B, Xia N, Sukhwani M, Wang Z, 85. Møllgård K, Jespersen A, Lutterodt MC, Yding
The migration and loss of human primordial germ Carey CC, Mazurie A, Cui J, Wilkinson R, Andersen C, Høyer PE, Byskov AG. Human pri-
stem cells from the hind gut epithelium towards Wiedenheft B, Irie N, Surani MA, Orwig KE, Reijo mordial germ cells migrate along nerve fibers and
the gonadal ridge. Int J Dev Biol. 2012;56(10–12): Pera RA. A PAX5–OCT4–PRDM1 developmental Schwann cells from the dorsal hind gut mesentery
771–778. switch specifies human primordial germ cells. Nat to the gonadal ridge. Mol Hum Reprod. 2010;16(9):
55. Anderson RA, Fulton N, Cowan G, Coutts S, Cell Biol. 2018;20(6):655–665. 621–631.
Saunders PT. Conserved and divergent patterns of 69. Yokobayashi S, Okita K, Nakagawa M, Nakamura T, 86. Makabe S, Motta PM. Migration of human germ
expression of DAZL, VASA and OCT4 in the germ Yabuta Y, Yamamoto T, Saitou M. Clonal variation cells and their relationship with the developing
cells of the human fetal ovary and testis. BMC Dev of human induced pluripotent stem cells for in- ovary: ultrastructural aspects. Prog Clin Biol Res.
Biol. 2007;7(1):136. duction into the germ cell fate. Biol Reprod. 2017; 1989;296:41–54.
56. Perrett RM, Turnpenny L, Eckert JJ, O’Shea M, Sonne 96(6):1154–1166. 87. Schneider DT, Schuster AE, Fritsch MK, Hu J, Olson
SB, Cameron IT, Wilson DI, Rajpert-De Meyts E, 70. Irie N, Tang WW, Azim Surani M. Germ cell T, Lauer S, Göbel U, Perlman EJ. Multipoint im-
Hanley NA. The early human germ cell lineage does specification and pluripotency in mammals: a printing analysis indicates a common precursor cell
not express SOX2 during in vivo development or perspective from early embryogenesis. Reprod Med for gonadal and nongonadal pediatric germ cell
upon in vitro culture. Biol Reprod. 2008;78(5): Biol. 2014;13(4):203–215. tumors. Cancer Res. 2001;61(19):7268–7276.
852–858. 71. Hackett JA, Surani MA. Regulatory principles of 88. Arora RS, Alston RD, Eden TO, Geraci M, Birch JM.
57. Aeckerle N, Drummer C, Debowski K, Viebahn C, pluripotency: from the ground state up. Cell Stem Comparative incidence patterns and trends of

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


Behr R. Primordial germ cell development in the Cell. 2014;15(4):416–430. gonadal and extragonadal germ cell tumors in
marmoset monkey as revealed by pluripotency 72. Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans England, 1979 to 2003. Cancer. 2012;118(17):
factor expression: suggestion of a novel model of EP, Mack DL, Gardner RL, McKay RD. New cell lines 4290–4297.
embryonic germ cell translocation. Mol Hum from mouse epiblast share defining features with 89. Runyan C, Schaible K, Molyneaux K, Wang Z, Levin
Reprod. 2015;21(1):66–80. human embryonic stem cells. Nature. 2007; L, Wylie C. Steel factor controls midline cell death of
58. Pereda J, Zorn T, Soto-Suazo M. Migration of hu- 448(7150):196–199. primordial germ cells and is essential for their
man and mouse primordial germ cells and colo- 73. Brons IG, Smithers LE, Trotter MW, Rugg-Gunn P, normal proliferation and migration. Development.
nization of the developing ovary: an ultrastructural Sun B, Chuva de Sousa Lopes SM, Howlett SK, 2006;133(24):4861–4869.
and cytochemical study. Microsc Res Tech. 2006; Clarkson A, Ahrlund-Richter L, Pedersen RA, Vallier 90. Gu Y, Runyan C, Shoemaker A, Surani A, Wylie C.
69(6):386–395. L. Derivation of pluripotent epiblast stem cells from Steel factor controls primordial germ cell survival
59. Fujimoto T, Miyayama Y, Fuyuta M. The origin, mammalian embryos. Nature. 2007;448(7150): and motility from the time of their specification in
migration and fine morphology of human pri- 191–195. the allantois, and provides a continuous niche
mordial germ cells. Anat Rec. 1977;188(3):315–330. 74. Hayashi K, Surani MA. Self-renewing epiblast stem throughout their migration. Development. 2009;
60. Kee K, Angeles VT, Flores M, Nguyen HN, Reijo Pera 136(8):1295–1303.
cells exhibit continual delineation of germ cells with
RA. Human DAZL, DAZ and BOULE genes modulate 91. Upadhyay S, Zamboni L. Ectopic germ cells: natural
epigenetic reprogramming in vitro. Development.
primordial germ-cell and haploid gamete forma- model for the study of germ cell sexual differen-
2009;136(21):3549–3556.
tion. Nature. 2009;462(7270):222–225. tiation. Proc Natl Acad Sci USA. 1982;79(21):
75. Sugawa F, Araúzo-Bravo MJ, Yoon J, Kim KP,
61. Irie N, Weinberger L, Tang WW, Kobayashi T, 6584–6588.
Aramaki S, Wu G, Stehling M, Psathaki OE, Hübner
Viukov S, Manor YS, Dietmann S, Hanna JH, Surani 92. Samokhvalov IM. Return to the hematopoietic
K, Schöler HR. Human primordial germ cell com-
MA. SOX17 is a critical specifier of human pri- stem cell origin. Cell Regen (Lond). 2012;1(1):9.
mitment in vitro associates with a unique PRDM14
mordial germ cell fate. Cell. 2015;160(1–2):253–268. 93. Fujimoto T, Yoshinaga K, Kono I. Distribution of
expression profile. EMBO J. 2015;34(8):1009–1024.
62. Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, fibronectin on the migratory pathway of primordial
76. Gell JJ, Zhao J, Chen D, Hunt TJ, Clark AT. PRDM14 is
Yabuta Y, Kurimoto K, Ohta H, Moritoki Y, Iwatani germ cells in mice. Anat Rec. 1985;211(3):271–278.
expressed in germ cell tumors with constitutive
C, Tsuchiya H, Nakamura S, Sekiguchi K, Sakuma T, 94. De Felici M, Scaldaferri ML, Farini D. Adhesion
overexpression altering human germline differen-
Yamamoto T, Mori T, Woltjen K, Nakagawa M, molecules for mouse primordial germ cells. Front
Yamamoto T, Takahashi K, Yamanaka S, Saitou M. tiation and proliferation. Stem Cell Res (Amst). 2018; Biosci. 2005;10(1–3):542–551.
Robust in vitro induction of human germ cell fate 27:46–56. 95. Richardson BE, Lehmann R. Mechanisms guiding
from pluripotent stem cells. Cell Stem Cell. 2015; 77. Barton LJ, LeBlanc MG, Lehmann R. Finding their primordial germ cell migration: strategies from
17(2):178–194. way: themes in germ cell migration. Curr Opin Cell different organisms. Nat Rev Mol Cell Biol. 2010;
63. Irie N, Surani MA. Efficient induction and isolation Biol. 2016;42:128–137. 11(1):37–49.
of human primordial germ cell-like cells from 78. Freeman B. The active migration of germ cells in the 96. Li L, Dong J, Yan L, Yong J, Liu X, Hu Y, Fan X, Wu X,
competent human pluripotent stem cells. Methods embryos of mice and men is a myth. Reproduction. Guo H, Wang X, Zhu X, Li R, Yan J, Wei Y, Zhao Y,
Mol Biol. 2017;1463:217–226. 2003;125(5):635–643. Wang W, Ren Y, Yuan P, Yan Z, Hu B, Guo F, Wen L,
64. Kanai-Azuma M, Kanai Y, Gad JM, Tajima Y, Taya C, 79. Kuwana T, Fujimoto T. Active locomotion of hu- Tang F, Qiao J. Single-cell RNA-seq analysis maps
Kurohmaru M, Sanai Y, Yonekawa H, Yazaki K, Tam man primordial germ cells in vitro. Anat Rec. 1983; development of human germline cells and gonadal
PP, Hayashi Y. Depletion of definitive gut endoderm 205(1):21–26. niche interactions [published correction appears in
in Sox17-null mutant mice. Development. 2002; 80. Donovan PJ, Stott D, Cairns LA, Heasman J, Wylie Cell Stem Cell. 2017;20(6):891–892]. Cell Stem Cell.
129(10):2367–2379. CC. Migratory and postmigratory mouse primordial 2017;20(6):858–873.e4.
65. Hara K, Kanai-Azuma M, Uemura M, Shitara H, Taya germ cells behave differently in culture. Cell. 1986; 97. Hamer G, de Rooij DG. Mutations causing specific
C, Yonekawa H, Kawakami H, Tsunekawa N, 44(6):831–838. arrests in the development of mouse primordial
Kurohmaru M, Kanai Y. Evidence for crucial role of 81. Godin I, Wylie C, Heasman J. Genital ridges exert germ cells and gonocytes. Biol Reprod. 2018;99(1):
hindgut expansion in directing proper migration of long-range effects on mouse primordial germ cell 75–86.
primordial germ cells in mouse early embryogenesis. numbers and direction of migration in culture. 98. Anderson R, Copeland TK, Schöler H, Heasman J,
Dev Biol. 2009;330(2):427–439. Development. 1990;108(2):357–363. Wylie C. The onset of germ cell migration in the
66. Kojima Y, Sasaki K, Yokobayashi S, Sakai Y, 82. Molyneaux KA, Stallock J, Schaible K, Wylie C. Time- mouse embryo. Mech Dev. 2000;91(1–2):61–68.
Nakamura T, Yabuta Y, Nakaki F, Nagaoka S, lapse analysis of living mouse germ cell migration. 99. Ara T, Nakamura Y, Egawa T, Sugiyama T, Abe K,
Woltjen K, Hotta A, Yamamoto T, Saitou M. Dev Biol. 2001;240(2):488–498. Kishimoto T, Matsui Y, Nagasawa T. Impaired
Evolutionarily distinctive transcriptional and sig- 83. Molyneaux KA, Zinszner H, Kunwar PS, Schaible K, colonization of the gonads by primordial germ cells
naling programs drive human germ cell lineage Stebler J, Sunshine MJ, O’Brien W, Raz E, Littman D, in mice lacking a chemokine, stromal cell-derived
specification from pluripotent stem cells. Cell Stem Wylie C, Lehmann R. The chemokine SDF1/CXCL12 factor-1 (SDF-1). Proc Natl Acad Sci USA. 2003;
Cell. 2017;21(4):517–532.e5. and its receptor CXCR4 regulate mouse germ cell 100(9):5319–5323.
67. Kobayashi T, Zhang H, Tang WWC, Irie N, Withey S, migration and survival. Development. 2003;130(18): 100. McCoshen JA, McCallion DJ. A study of the pri-
Klisch D, Sybirna A, Dietmann S, Contreras DA, 4279–4286. mordial germ cells during their migratory phase in
Webb R, Allegrucci C, Alberio R, Surani MA. Prin- 84. Wrobel KH, Süss F. Identification and tempor- Steel mutant mice. Experientia. 1975;31(5):589–590.
ciples of early human development and germ cell ospatial distribution of bovine primordial germ cells 101. Gu Y, Runyan C, Shoemaker A, Surani MA, Wylie C.
program from conserved model systems. Nature. prior to gonadal sexual differentiation. Anat Membrane-bound steel factor maintains a high
2017;546(7658):416–420. Embryol (Berl). 1998;197(6):451–467. local concentration for mouse primordial germ cell

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 895


REVIEW

motility, and defines the region of their migration. 118. Toyooka Y, Tsunekawa N, Takahashi Y, Matsui Y, reversal with partial duplication of chromosome
PLoS One. 2011;6(10):e25984. Satoh M, Noce T. Expression and intracellular arm 22q. Am J Med Genet A. 2004;127A(2):149–151.
102. Matsui Y, Zsebo KM, Hogan BL. Embryonic ex- localization of mouse Vasa-homologue protein 135. Sutton E, Hughes J, White S, Sekido R, Tan J,
pression of a haematopoietic growth factor enco- during germ cell development. Mech Dev. 2000; Arboleda V, Rogers N, Knower K, Rowley L, Eyre H,
ded by the SI locus and the ligand for c-kit. Nature. 93(1–2):139–149. Rizzoti K, McAninch D, Goncalves J, Slee J, Turbitt E,
1990;347(6294):667–669. 119. Seligman J, Page DC. The Dazh gene is expressed in Bruno D, Bengtsson H, Harley V, Vilain E, Sinclair A,
103. Keshet E, Lyman SD, Williams DE, Anderson DM, male and female embryonic gonads before germ Lovell-Badge R, Thomas P. Identification of SOX3 as
Jenkins NA, Copeland NG, Parada LF. Embryonic cell sex differentiation. Biochem Biophys Res Com- an XX male sex reversal gene in mice and humans.
RNA expression patterns of the c-kit receptor and mun. 1998;245(3):878–882. J Clin Invest. 2011;121(1):328–341.
its cognate ligand suggest multiple functional roles 120. Enders GC, May JJ II. Developmentally regulated 136. Karl J, Capel B. Sertoli cells of the mouse testis
in mouse development. EMBO J. 1991;10(9): expression of a mouse germ cell nuclear antigen originate from the coelomic epithelium. Dev Biol.
2425–2435. examined from embryonic day 11 to adult in male 1998;203(2):323–333.
104. Laird DJ, Altshuler-Keylin S, Kissner MD, Zhou X, and female mice. Dev Biol. 1994;163(2):331–340. 137. Schmahl J, Eicher EM, Washburn LL, Capel B. Sry
Anderson KV. Ror2 enhances polarity and di- 121. Chang DH, Cattoretti G, Calame KL. The dynamic induces cell proliferation in the mouse gonad.
rectional migration of primordial germ cells. PLoS expression pattern of B lymphocyte induced Development. 2000;127(1):65–73.
Genet. 2011;7(12):e1002428. maturation protein-1 (Blimp-1) during mouse 138. Liu C, Rodriguez K, Yao HH. Mapping lineage
105. Arora R, Altman E, Tran ND, Laird DJ. Novel do- embryonic development. Mech Dev. 2002;117(1–2): progression of somatic progenitor cells in the

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


mains of expression for orphan receptor tyrosine 305–309. mouse fetal testis. Development. 2016;143(20):
kinase Ror2 in the human and mouse reproductive 122. Cooke JE, Godin I, Ffrench-Constant C, Heasman J, 3700–3710.
system. Dev Dyn. 2014;243(8):1037–1045. Wylie CC. Culture and manipulation of primordial 139. Armstrong JF, Pritchard-Jones K, Bickmore WA,
106. Bendel-Stenzel MR, Gomperts M, Anderson R, germ cells. Methods Enzymol. 1993;225:37–58. Hastie ND, Bard JB. The expression of the Wilms’
Heasman J, Wylie C. The role of cadherins during 123. Coucouvanis EC, Jones PP. Changes in proto- tumour gene, WT1, in the developing mammalian
primordial germ cell migration and early gonad oncogene expression correlated with general and embryo. Mech Dev. 1993;40(1–2):85–97.
formation in the mouse. Mech Dev. 2000;91(1–2): sex-specific differentiation in murine primordial 140. Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock
143–152. germ cells. Mech Dev. 1993;42(1–2):49–58. D, Ziegler U, Gubler MC, Schedl A. Two splice
107. Di Carlo A, De Felici M. A role for E-cadherin in 124. Manova K, Bachvarova RF. Expression of c-kit variants of the Wilms’ tumor 1 gene have distinct
mouse primordial germ cell development. Dev Biol. encoded at the W locus of mice in developing functions during sex determination and nephron
2000;226(2):209–219. embryonic germ cells and presumptive melano- formation. Cell. 2001;106(3):319–329.
108. Anderson R, Fässler R, Georges-Labouesse E, Hynes blasts. Dev Biol. 1991;146(2):312–324. 141. Hu YC, Okumura LM, Page DC. Gata4 is required for
RO, Bader BL, Kreidberg JA, Schaible K, Heasman J, 125. Gkountela S, Zhang KX, Shafiq TA, Liao WW, formation of the genital ridge in mice. PLoS Genet.
Wylie C. Mouse primordial germ cells lacking beta1 Hargan-Calvopiña J, Chen PY, Clark AT. DNA 2013;9(7):e1003629.
integrins enter the germline but fail to migrate demethylation dynamics in the human prenatal 142. Miyamoto N, Yoshida M, Kuratani S, Matsuo I,
normally to the gonads. Development. 1999;126(8): germline. Cell. 2015;161(6):1425–1436. Aizawa S. Defects of urogenital development in
1655–1664. 126. Hanley NA, Hagan DM, Clement-Jones M, Ball SG, mice lacking Emx2. Development. 1997;124(9):
109. Alvarez-Buylla A, Merchant-Larios H. Mouse pri- Strachan T, Salas-Cortés L, McElreavey K, Lindsay S, 1653–1664.
mordial germ cells use fibronectin as a substrate for Robson S, Bullen P, Ostrer H, Wilson DI. SRY, SOX9, 143. Birk OS, Casiano DE, Wassif CA, Cogliati T, Zhao L,
migration. Exp Cell Res. 1986;165(2):362–368. and DAX1 expression patterns during human sex Zhao Y, Grinberg A, Huang S, Kreidberg JA, Parker
110. Chuva de Sousa Lopes SM, van den Driesche S, determination and gonadal development. Mech KL, Porter FD, Westphal H. The LIM homeobox
Carvalho RL, Larsson J, Eggen B, Surani MA, Dev. 2000;91(1–2):403–407. gene Lhx9 is essential for mouse gonad formation.
Mummery CL. Altered primordial germ cell mi- 127. Miller WL, Auchus RJ. The molecular biology, bio- Nature. 2000;403(6772):909–913.
gration in the absence of transforming growth chemistry, and physiology of human steroidogen- 144. Luo X, Ikeda Y, Parker KL. A cell-specific nuclear
factor b signaling via ALK5. Dev Biol. 2005;284(1): esis and its disorders. Endocr Rev. 2011;32(1):81–151. receptor is essential for adrenal and gonadal de-
194–203. 128. Ahmed SF, Bashamboo A, Lucas-Herald A, velopment and sexual differentiation. Cell. 1994;
111. Mattiske D, Kume T, Hogan BL. The mouse fork- McElreavey K. Understanding the genetic aetiology 77(4):481–490.
head gene Foxc1 is required for primordial germ cell in patients with XY DSD. Br Med Bull. 2013;106(1): 145. Wilhelm D, Englert C. The Wilms tumor suppressor
migration and antral follicle development. Dev Biol. 67–89. WT1 regulates early gonad development by acti-
2006;290(2):447–458. 129. Bashamboo A, Eozenou C, Rojo S, McElreavey K. vation of Sf1. Genes Dev. 2002;16(14):1839–1851.
112. Takeuchi Y, Molyneaux K, Runyan C, Schaible K, Anomalies in human sex determination provide 146. Hanley NA, Ball SG, Clement-Jones M, Hagan DM,
Wylie C. The roles of FGF signaling in germ cell unique insights into the complex genetic in- Strachan T, Lindsay S, Robson S, Ostrer H, Parker KL,
migration in the mouse. Development. 2005;132(24): teractions of early gonad development. Clin Genet. Wilson DI. Expression of steroidogenic factor 1 and
5399–5409. 2017;91(2):143–156. Wilms’ tumour 1 during early human gonadal
113. Clermont Y, Perey B. Quantitative study of the cell 130. Koopman P, Gubbay J, Vivian N, Goodfellow P, development and sex determination. Mech Dev.
population of the seminiferous tubules in immature Lovell-Badge R. Male development of chromoso- 1999;87(1-2):175–180.
rats. Am J Anat. 1957;100(2):241–267. mally female mice transgenic for Sry. Nature. 1991; 147. Kusaka M, Katoh-Fukui Y, Ogawa H, Miyabayashi K,
114. McKinnell C, Mitchell RT, Morris K, Anderson RA, 351(6322):117–121. Baba T, Shima Y, Sugiyama N, Sugimoto Y, Okuno Y,
Kelnar CJ, Wallace WH, Sharpe RM. Perinatal germ 131. Foster JW, Dominguez-Steglich MA, Guioli S, Kwok Kodama R, Iizuka-Kogo A, Senda T, Sasaoka T,
cell development and differentiation in the male C, Weller PA, Stevanović M, Weissenbach J, Kitamura K, Aizawa S, Morohashi K. Abnormal
marmoset (Callithrix jacchus): similarities with the Mansour S, Young ID, Goodfellow PN, Brook JD, epithelial cell polarity and ectopic epidermal
human and differences from the rat. Hum Reprod. Schafer AJ. Campomelic dysplasia and autosomal growth factor receptor (EGFR) expression induced
2013;28(4):886–896. sex reversal caused by mutations in an SRY-related in Emx2 KO embryonic gonads. Endocrinology. 2010;
115. Hara K, Nakagawa T, Enomoto H, Suzuki M, gene. Nature. 1994;372(6506):525–530. 151(12):5893–5904.
Yamamoto M, Simons BD, Yoshida S. Mouse 132. Wagner T, Wirth J, Meyer J, Zabel B, Held M, 148. Katoh-Fukui Y, Miyabayashi K, Komatsu T, Owaki A,
spermatogenic stem cells continually interconvert Zimmer J, Pasantes J, Bricarelli FD, Keutel J, Hustert E, Baba T, Shima Y, Kidokoro T, Kanai Y, Schedl A,
between equipotent singly isolated and syncytial Wolf U, Tommerup N, Schempp W, Scherer G. Wilhelm D, Koopman P, Okuno Y, Morohashi K.
states. Cell Stem Cell. 2014;14(5):658–672. Autosomal sex reversal and campomelic dysplasia Cbx2, a polycomb group gene, is required for Sry
116. Yoshida S, Sukeno M, Nabeshima Y. A vasculature- are caused by mutations in and around the SRY- gene expression in mice. Endocrinology. 2012;153(2):
associated niche for undifferentiated spermatogo- related gene SOX9. Cell. 1994;79(6):1111–1120. 913–924.
nia in the mouse testis. Science. 2007;317(5845): 133. Morais da Silva S, Hacker A, Harley V, Goodfellow P, 149. Katoh-Fukui Y, Tsuchiya R, Shiroishi T, Nakahara Y,
1722–1726. Swain A, Lovell-Badge R. Sox9 expression during Hashimoto N, Noguchi K, Higashinakagawa T.
117. Molyneaux KA, Wang Y, Schaible K, Wylie C. gonadal development implies a conserved role for Male-to-female sex reversal in M33 mutant mice.
Transcriptional profiling identifies genes differen- the gene in testis differentiation in mammals and Nature. 1998;393(6686):688–692.
tially expressed during and after migration in birds. Nat Genet. 1996;14(1):62–68. 150. Fujimoto Y, Tanaka SS, Yamaguchi YL, Kobayashi H,
murine primordial germ cells. Gene Expr Patterns. 134. Seeherunvong T, Perera EM, Bao Y, Benke PJ, Kuroki S, Tachibana M, Shinomura M, Kanai Y,
2004;4(2):167–181. Benigno A, Donahue RP, Berkovitz GD. 46,XX sex Morohashi K, Kawakami K, Nishinakamura R.

896 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Homeoproteins Six1 and Six4 regulate male sex requirement for MAPK signalling in mouse sex modulation of Sf-1 dosage. Development. 2007;
determination and mouse gonadal development. determination. PLoS Biol. 2009;7(9):e1000196. 134(12):2349–2358.
Dev Cell. 2013;26(4):416–430. 166. Pearlman A, Loke J, Le Caignec C, White S, Chin L, 182. Combes AN, Spiller CM, Harley VR, Sinclair AH,
151. Schnabel CA, Selleri L, Cleary ML. Pbx1 is essential Friedman A, Warr N, Willan J, Brauer D, Farmer C, Dunwoodie SL, Wilhelm D, Koopman P. Gonadal
for adrenal development and urogenital differen- Brooks E, Oddoux C, Riley B, Shajahan S, Camerino defects in Cited2-mutant mice indicate a role for
tiation. Genesis. 2003;37(3):123–130. G, Homfray T, Crosby AH, Couper J, David A, SF1 in both testis and ovary differentiation. Int J Dev
152. Pitetti JL, Calvel P, Zimmermann C, Conne B, Greenfield A, Sinclair A, Ostrer H. Mutations in Biol. 2010;54(4):683–689.
Papaioannou MD, Aubry F, Cederroth CR, Urner F, MAP3K1 cause 46,XY disorders of sex development 183. Buaas FW, Val P, Swain A. The transcription co-
Fumel B, Crausaz M, Docquier M, Herrera PL, and implicate a common signal transduction factor CITED2 functions during sex determination
Pralong F, Germond M, Guillou F, Jégou B, Nef S. An pathway in human testis determination. Am J Hum and early gonad development. Hum Mol Genet.
essential role for insulin and IGF1 receptors in Genet. 2010;87(6):898–904. 2009;18(16):2989–3001.
regulating sertoli cell proliferation, testis size, and 167. Warr N, Bogani D, Siggers P, Brixey R, Tateossian H, 184. Hossain A, Saunders GF. The human sex-
FSH action in mice. Mol Endocrinol. 2013;27(5): Dopplapudi A, Wells S, Cheeseman M, Xia Y, Ostrer determining gene SRY is a direct target of WT1.
814–827. H, Greenfield A. Minor abnormalities of testis de- J Biol Chem. 2001;276(20):16817–16823.
153. Koopman P, Münsterberg A, Capel B, Vivian N, velopment in mice lacking the gene encoding the 185. Bor YC, Swartz J, Morrison A, Rekosh D, Ladomery
Lovell-Badge R. Expression of a candidate sex- MAPK signalling component, MAP3K1. PLoS One. M, Hammarskjöld ML. The Wilms’ tumor 1 (WT1)
determining gene during mouse testis differentia- 2011;6(5):e19572. gene (1KTS isoform) functions with a CTE to

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


tion. Nature. 1990;348(6300):450–452. 168. Nef S, Verma-Kurvari S, Merenmies J, Vassalli JD, enhance translation from an unspliced RNA with a
154. Hacker A, Capel B, Goodfellow P, Lovell-Badge R. Efstratiadis A, Accili D, Parada LF. Testis de- retained intron. Genes Dev. 2006;20(12):1597–1608.
Expression of Sry, the mouse sex determining gene. termination requires insulin receptor family func- 186. Shimamura R, Fraizer GC, Trapman J, Lau YfC,
Development. 1995;121(6):1603–1614. tion in mice. Nature. 2003;426(6964):291–295. Saunders GF. The Wilms’ tumor gene WT1 can
155. Jeske YW, Bowles J, Greenfield A, Koopman P. Ex- 169. Miyamoto Y, Taniguchi H, Hamel F, Silversides DW, regulate genes involved in sex determination and
pression of a linear Sry transcript in the mouse Viger RS. A GATA4/WT1 cooperation regulates differentiation: SRY, Müllerian-inhibiting substance,
genital ridge. Nat Genet. 1995;10(4):480–482. transcription of genes required for mammalian sex and the androgen receptor. Clin Cancer Res. 1997;
156. Del Valle I, Buonocore F, Duncan AJ, Lin L, Barenco determination and differentiation. BMC Mol Biol. 3(12 Pt 2):2571–2580.
M, Parnaik R, Shah S, Hubank M, Gerrelli D, 2008;9:44. 187. Matsuzawa-Watanabe Y, Inoue J, Semba K. Tran-
Achermann JC. A genomic atlas of human adrenal 170. Zhang Y, Ai F, Zheng J, Peng B. Associations of scriptional activity of testis-determining factor SRY
and gonad development. Wellcome Open Res. 2017; GATA4 genetic mutations with the risk of con- is modulated by the Wilms’ tumor 1 gene product,
2:25. genital heart disease: a meta-analysis. Medicine WT1. Oncogene. 2003;22(39):7900–7904.
157. Bullejos M, Koopman P. Spatially dynamic expres- (Baltimore). 2017;96(18):e6857. 188. Hoyle C, Narvaez V, Alldus G, Lovell-Badge R, Swain
sion of Sry in mouse genital ridges. Dev Dyn. 2001; 171. Ostrer H, Huang HY, Masch RJ, Shapiro E. A cellular A. Dax1 expression is dependent on steroidogenic
study of human testis development. Sex Dev. 2007; factor 1 in the developing gonad. Mol Endocrinol.
221(2):201–205.
1(5):286–292. 2002;16(4):747–756.
158. Hiramatsu R, Matoba S, Kanai-Azuma M,
172. de Santa Barbara P, Méjean C, Moniot B, Malclès 189. Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC,
Tsunekawa N, Katoh-Fukui Y, Kurohmaru M,
MH, Berta P, Boizet-Bonhoure B. Steroidogenic Tonini G, Ferrante E, Chiumello G, McCabe ER,
Morohashi K, Wilhelm D, Koopman P, Kanai Y. A
factor-1 contributes to the cyclic-adenosine Fraccaro M, Zuffardi O, Camerino G. A dosage
critical time window of Sry action in gonadal sex
monophosphate down-regulation of human SRY sensitive locus at chromosome Xp21 is involved in
determination in mice. Development. 2009;136(1):
gene expression. Biol Reprod. 2001;64(3):775–783. male to female sex reversal. Nat Genet. 1994;7(4):
129–138.
173. Achermann JC, Ito M, Ito M, Hindmarsh PC, 497–501.
159. Wilhelm D, Washburn LL, Truong V, Fellous M,
Jameson JL. A mutation in the gene encoding 190. Swain A, Narvaez V, Burgoyne P, Camerino G,
Eicher EM, Koopman P. Antagonism of the testis-
steroidogenic factor-1 causes XY sex reversal and Lovell-Badge R. Dax1 antagonizes Sry action in
and ovary-determining pathways during ovotestis
adrenal failure in humans. Nat Genet. 1999;22(2): mammalian sex determination. Nature. 1998;
development in mice. Mech Dev. 2009;126(5–6):
125–126. 391(6669):761–767.
324–336. 174. Parker KL, Rice DA, Lala DS, Ikeda Y, Luo X, Wong M, 191. Ludbrook LM, Bernard P, Bagheri-Fam S, Ryan J,
160. Larney C, Bailey TL, Koopman P. Switching on sex:
Bakke M, Zhao L, Frigeri C, Hanley NA, Stallings N, Sekido R, Wilhelm D, Lovell-Badge R, Harley VR.
transcriptional regulation of the testis-determining Schimmer BP. Steroidogenic factor 1: an essential Excess DAX1 leads to XY ovotesticular disorder of
gene Sry. Development. 2014;141(11):2195–2205. mediator of endocrine development. Recent Prog sex development (DSD) in mice by inhibiting ste-
161. Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Horm Res. 2002;57(1):19–36. roidogenic factor-1 (SF1) activation of the testis
Eicher EM, Orkin SH. Gonadal differentiation, sex 175. El-Khairi R, Achermann JC. Steroidogenic factor-1 enhancer of SRY-box-9 (Sox9). Endocrinology. 2012;
determination and normal Sry expression in mice and human disease. Semin Reprod Med. 2012;30(5): 153(4):1948–1958.
require direct interaction between transcription 374–381. 192. Barbaro M, Oscarson M, Schoumans J, Staaf J,
partners GATA4 and FOG2. Development. 2002; 176. Domenice S, Machado AZ, Ferreira FM, Ferraz-de- Ivarsson SA, Wedell A. Isolated 46,XY gonadal
129(19):4627–4634. Souza B, Lerario AM, Lin L, Nishi MY, Gomes NL, da dysgenesis in two sisters caused by a Xp21.2 in-
162. Manuylov NL, Zhou B, Ma Q, Fox SC, Pu WT, Silva TE, Silva RB, Correa RV, Montenegro LR, terstitial duplication containing the DAX1 gene.
Tevosian SG. Conditional ablation of Gata4 and Narciso A, Costa EM, Achermann JC, Mendonca BB. J Clin Endocrinol Metab. 2007;92(8):3305–3313.
Fog2 genes in mice reveals their distinct roles in Wide spectrum of NR5A1-related phenotypes in 193. Bouma GJ, Albrecht KH, Washburn LL, Recknagel
mammalian sexual differentiation. Dev Biol. 2011; 46,XY and 46,XX individuals. Birth Defects Res C AK, Churchill GA, Eicher EM. Gonadal sex reversal in
353(2):229–241. Embryo Today. 2016;108(4):309–320. mutant Dax1 XY mice: a failure to upregulate Sox9
163. Gierl MS, Gruhn WH, von Seggern A, Maltry N, 177. Ikeda Y, Shen WH, Ingraham HA, Parker KL. De- in pre-Sertoli cells. Development. 2005;132(13):
Niehrs C. GADD45G functions in male sex de- velopmental expression of mouse steroidogenic 3045–3054.
termination by promoting p38 signaling and Sry factor-1, an essential regulator of the steroid hy- 194. Yu RN, Ito M, Saunders TL, Camper SA, Jameson JL.
expression. Dev Cell. 2012;23(5):1032–1042. droxylases. Mol Endocrinol. 1994;8(5):654–662. Role of Ahch in gonadal development and game-
164. Warr N, Carre GA, Siggers P, Faleato JV, Brixey R, 178. Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C, togenesis. Nat Genet. 1998;20(4):353–357.
Pope M, Bogani D, Childers M, Wells S, Scudamore Schoenle EJ. Ovaries and female phenotype in a girl 195. Meeks JJ, Crawford SE, Russell TA, Morohashi K,
CL, Tedesco M, del Barco Barrantes I, Nebreda AR, with 46,XY karyotype and mutations in the CBX2 Weiss J, Jameson JL. Dax1 regulates testis cord or-
Trainor PA, Greenfield A. Gadd45g and Map3k4 gene. Am J Hum Genet. 2009;84(5):658–663. ganization during gonadal differentiation. Devel-
interactions regulate mouse testis determination 179. Gil J, O’Loghlen A. PRC1 complex diversity: where is opment. 2003;130(5):1029–1036.
via p38 MAPK-mediated control of Sry expression. it taking us? Trends Cell Biol. 2014;24(11):632–641. 196. Park SY, Lee EJ, Emge D, Jahn CL, Jameson JL. A
Dev Cell. 2012;23(5):1020–1031. 180. Eid W, Opitz L, Biason-Lauber A. Genome-wide phenotypic spectrum of sexual development in
165. Bogani D, Siggers P, Brixey R, Warr N, Beddow S, identification of CBX2 targets: insights in the hu- Dax1 (Nr0b1)-deficient mice: consequence of the
Edwards J, Williams D, Wilhelm D, Koopman P, man sex development network. Mol Endocrinol. C57BL/6J strain on sex determination. Biol Reprod.
Flavell RA, Chi H, Ostrer H, Wells S, Cheeseman M, 2015;29(2):247–257. 2008;79(6):1038–1045.
Greenfield A. Loss of mitogen-activated pro- 181. Val P, Martinez-Barbera JP, Swain A. Adrenal de- 197. Swain A, Zanaria E, Hacker A, Lovell-Badge R,
tein kinase kinase kinase 4 (MAP3K4) reveals a velopment is initiated by Cited2 and Wt1 through Camerino G. Mouse Dax1 expression is consistent

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 897


REVIEW

with a role in sex determination as well as in adrenal not a common cause of 46,XX testicular or 46,XX 231. Georg I, Bagheri-Fam S, Knower KC, Wieacker P,
and hypothalamus function. Nat Genet. 1996;12(4): ovotesticular DSD. J Pediatr Endocrinol Metab. 2012; Scherer G, Harley VR. Mutations of the SRY-
404–409. 25(1-2):121–123. responsive enhancer of SOX9 are uncommon in
198. Ludbrook LM, Harley VR. Sex determination: a 217. Refai O, Friedman A, Terry L, Jewett T, Pearlman A, XY gonadal dysgenesis. Sex Dev. 2010;4(6):321–325.
“window” of DAX1 activity. Trends Endocrinol Perle MA, Ostrer H. De novo 12;17 translocation 232. Aksglaede L, Sørensen K, Boas M, Mouritsen A,
Metab. 2004;15(3):116–121. upstream of SOX9 resulting in 46,XX testicular Hagen CP, Jensen RB, Petersen JH, Linneberg A,
199. Nishino K, Hattori N, Tanaka S, Shiota K. DNA disorder of sex development. Am J Med Genet A. Andersson AM, Main KM, Skakkebæk NE, Juul A.
methylation-mediated control of Sry gene expres- 2010;152A(2):422–426. Changes in anti-Müllerian hormone (AMH)
sion in mouse gonadal development. J Biol Chem. 218. Xiao B, Ji X, Xing Y, Chen YW, Tao J. A rare case of throughout the life span: a population-based study
2004;279(21):22306–22313. 46,XX SRY-negative male with approximately 74-kb of 1027 healthy males from birth (cord blood) to
200. Dasari VK, Deng D, Perinchery G, Yeh CC, Dahiya R. duplication in a region upstream of SOX9. Eur J Med the age of 69 years. J Clin Endocrinol Metab. 2010;
DNA methylation regulates the expression of Y Genet. 2013;56(12):695–698. 95(12):5357–5364.
chromosome specific genes in prostate cancer. 219. Benko S, Gordon CT, Mallet D, Sreenivasan R, 233. Zhou B, Hutson JM. Human chorionic gonado-
J Urol. 2002;167(1):335–338. Thauvin-Robinet C, Brendehaug A, Thomas S, tropin (hCG) fails to stimulate gonocyte differen-
201. Kuroki S, Matoba S, Akiyoshi M, Matsumura Y, Bruland O, David M, Nicolino M, Labalme A, tiation in newborn mouse testes in organ culture.
Miyachi H, Mise N, Abe K, Ogura A, Wilhelm D, Sanlaville D, Callier P, Malan V, Huet F, Molven A, J Urol. 1995;153(2):501–505.
Koopman P, Nozaki M, Kanai Y, Shinkai Y, Dijoud F, Munnich A, Faivre L, Amiel J, Harley V, 234. Zhou B, Watts LM, Hutson JM. Germ cell devel-

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


Tachibana M. Epigenetic regulation of mouse sex Houge G, Morel Y, Lyonnet S. Disruption of a long opment in neonatal mouse testes in vitro requires
determination by the histone demethylase Jmjd1a. distance regulatory region upstream of SOX9 in Müllerian inhibiting substance. J Urol. 1993;150(2 Pt
Science. 2013;341(6150):1106–1109. isolated disorders of sex development. J Med Genet. 2):613–616.
202. Carré GA, Siggers P, Xipolita M, Brindle P, Lutz B, 2011;48(12):825–830. 235. Josso N, Rey RA, Picard JY. Anti-Müllerian hormone:
Wells S, Greenfield A. Loss of p300 and CBP disrupts 220. Chaboissier MC, Kobayashi A, Vidal VI, Lützkendorf a valuable addition to the toolbox of the pediatric
histone acetylation at the mouse Sry promoter and S, van de Kant HJ, Wegner M, de Rooij DG, endocrinologist. Int J Endocrinol. 2013;2013:674105.
causes XY gonadal sex reversal. Hum Mol Genet. Behringer RR, Schedl A. Functional analysis of Sox8 236. Arango NA, Lovell-Badge R, Behringer RR. Targeted
2018;27(1):190–198. and Sox9 during sex determination in the mouse. mutagenesis of the endogenous mouse Mis gene
203. Kim Y, Kobayashi A, Sekido R, DiNapoli L, Brennan J, Development. 2004;131(9):1891–1901. promoter: in vivo definition of genetic pathways of
Chaboissier MC, Poulat F, Behringer RR, Lovell-Badge 221. Sekido R, Lovell-Badge R. Sex determination involves vertebrate sexual development. Cell. 1999;99(4):
R, Capel B. Fgf9 and Wnt4 act as antagonistic signals synergistic action of SRY and SF1 on a specific Sox9 409–419.
to regulate mammalian sex determination. PLoS Biol. enhancer. Nature. 2008;453(7197):930–934. 237. Giuili G, Shen WH, Ingraham HA. The nuclear re-
2006;4(6):e187. 222. Adams IR, McLaren A. Sexually dimorphic devel- ceptor SF-1 mediates sexually dimorphic expression
204. Rotgers E, Jørgensen A, Yao HH. At the crossroads of opment of mouse primordial germ cells: switching
of Mullerian inhibiting substance, in vivo. Devel-
fate—somatic cell lineage specification in the fetal from oogenesis to spermatogenesis. Development.
opment. 1997;124(9):1799–1807.
gonad. Endocr Rev. 2018;39(5):739–759. 2002;129(5):1155–1164.
238. Nachtigal MW, Hirokawa Y, Enyeart-VanHouten
205. Sekido R, Bar I, Narváez V, Penny G, Lovell-Badge R. 223. Wilhelm D, Martinson F, Bradford S, Wilson MJ,
DL, Flanagan JN, Hammer GD, Ingraham HA.
SOX9 is up-regulated by the transient expression of Combes AN, Beverdam A, Bowles J, Mizusaki H,
Wilms’ tumor 1 and Dax-1 modulate the orphan
SRY specifically in Sertoli cell precursors. Dev Biol. Koopman P. Sertoli cell differentiation is induced
nuclear receptor SF-1 in sex-specific gene expres-
2004;274(2):271–279. both cell-autonomously and through prostaglandin
sion. Cell. 1998;93(3):445–454.
206. Svingen T, Koopman P. Building the mammalian signaling during mammalian sex determination. Dev
239. Shen WH, Moore CC, Ikeda Y, Parker KL, Ingraham
testis: origins, differentiation, and assembly of the Biol. 2005;287(1):111–124.
HA. Nuclear receptor steroidogenic factor 1 regu-
component cell populations. Genes Dev. 2013; 224. Moniot B, Declosmenil F, Barrionuevo F, Scherer G,
lates the Müllerian inhibiting substance gene: a link
27(22):2409–2426. Aritake K, Malki S, Marzi L, Cohen-Solal A, Georg I,
to the sex determination cascade. Cell. 1994;77(5):
207. Palmer SJ, Burgoyne PS. In situ analysis of fetal, Klattig J, Englert C, Kim Y, Capel B, Eguchi N, Urade
651–661.
prepuberal and adult XX—XY chimaeric mouse Y, Boizet-Bonhoure B, Poulat F. The PGD2 pathway,
240. Tremblay JJ, Viger RS. Transcription factor GATA-4
testes: Sertoli cells are predominantly, but not ex- independently of FGF9, amplifies SOX9 activity in
clusively, XY. Development. 1991;112(1):265–268. Sertoli cells during male sexual differentiation. De- enhances Müllerian inhibiting substance gene
208. Colvin JS, Green RP, Schmahl J, Capel B, Ornitz DM. velopment. 2009;136(11):1813–1821. transcription through a direct interaction with the
Male-to-female sex reversal in mice lacking fibro- 225. Bagheri-Fam S, Sim H, Bernard P, Jayakody I, Taketo nuclear receptor SF-1. Mol Endocrinol. 1999;13(8):
blast growth factor 9. Cell. 2001;104(6):875–889. MM, Scherer G, Harley VR. Loss of Fgfr2 leads to 1388–1401.
209. Schmahl J, Capel B. Cell proliferation is necessary for partial XY sex reversal. Dev Biol. 2008;314(1):71–83. 241. Viger RS, Mertineit C, Trasler JM, Nemer M.
the determination of male fate in the gonad. Dev 226. Kim Y, Bingham N, Sekido R, Parker KL, Lovell-Badge Transcription factor GATA-4 is expressed in a
Biol. 2003;258(2):264–276. R, Capel B. Fibroblast growth factor receptor 2 sexually dimorphic pattern during mouse gonadal
210. Schmahl J, Kim Y, Colvin JS, Ornitz DM, Capel B. Fgf9 regulates proliferation and Sertoli differentiation development and is a potent activator of the
induces proliferation and nuclear localization of during male sex determination. Proc Natl Acad Sci Müllerian inhibiting substance promoter. Develop-
FGFR2 in Sertoli precursors during male sex de- USA. 2007;104(42):16558–16563. ment. 1998;125(14):2665–2675.
termination. Development. 2004;131(15):3627–3636. 227. Bagheri-Fam S, Ono M, Li L, Zhao L, Ryan J, Lai R, 242. Polanco JC, Wilhelm D, Davidson TL, Knight D,
211. Bullejos M, Koopman P. Delayed Sry and Sox9 Katsura Y, Rossello FJ, Koopman P, Scherer G, Koopman P. Sox10 gain-of-function causes XX sex
expression in developing mouse gonads underlies Bartsch O, Eswarakumar JV, Harley VR. FGFR2 reversal in mice: implications for human 22q-linked
B6-Y(DOM) sex reversal. Dev Biol. 2005;278(2): mutation in 46,XY sex reversal with craniosynos- disorders of sex development. Hum Mol Genet. 2010;
473–481. tosis. Hum Mol Genet. 2015;24(23):6699–6710. 19(3):506–516.
212. Hiramatsu R, Harikae K, Tsunekawa N, Kurohmaru 228. Malki S, Nef S, Notarnicola C, Thevenet L, Gasca S, 243. Nicol B, Yao HH. Gonadal identity in the absence of
M, Matsuo I, Kanai Y. FGF signaling directs a center- Méjean C, Berta P, Poulat F, Boizet-Bonhoure B. pro-testis factor SOX9 and pro-ovary factor beta-
to-pole expansion of tubulogenesis in mouse testis Prostaglandin D2 induces nuclear import of the sex- catenin in mice. Biol Reprod. 2015;93(2):35.
differentiation. Development. 2010;137(2):303–312. determining factor SOX9 via its cAMP-PKA 244. Yao HH, Whoriskey W, Capel B. Desert Hedgehog/
213. Huang B, Wang S, Ning Y, Lamb AN, Bartley J. phosphorylation. EMBO J. 2005;24(10):1798–1809. Patched 1 signaling specifies fetal Leydig cell fate in
Autosomal XX sex reversal caused by duplication of 229. Wilhelm D, Hiramatsu R, Mizusaki H, Widjaja L, testis organogenesis. Genes Dev. 2002;16(11):
SOX9. Am J Med Genet. 1999;87(4):349–353. Combes AN, Kanai Y, Koopman P. SOX9 regulates 1433–1440.
214. Vidal VP, Chaboissier MC, de Rooij DG, Schedl A. prostaglandin D synthase gene transcription in vivo 245. Vernet N, Dennefeld C, Rochette-Egly C, Oulad-
Sox9 induces testis development in XX transgenic to ensure testis development. J Biol Chem. 2007; Abdelghani M, Chambon P, Ghyselinck NB, Mark
mice. Nat Genet. 2001;28(3):216–217. 282(14):10553–10560. M. Retinoic acid metabolism and signaling path-
215. Cox JJ, Willatt L, Homfray T, Woods CGA. A SOX9 230. Matson CK, Murphy MW, Sarver AL, Griswold MD, ways in the adult and developing mouse testis.
duplication and familial 46,XX developmental tes- Bardwell VJ, Zarkower D. DMRT1 prevents female Endocrinology. 2006;147(1):96–110.
ticular disorder. N Engl J Med. 2011;364(1):91–93. reprogramming in the postnatal mammalian testis 246. Schepers G, Wilson M, Wilhelm D, Koopman P.
216. Seeherunvong T, Ukarapong S, McElreavey K, [published correction appears in Nature. 2011; SOX8 is expressed during testis differentiation in
Berkovitz GD, Perera EM. Duplication of SOX9 is 477(7363):238]. Nature. 2011;476(7358):101–104. mice and synergizes with SF1 to activate the Amh

898 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

promoter in vitro. J Biol Chem. 2003;278(30): 264. Buehr M, Gu S, McLaren A. Mesonephric contri- Granados J, Rosas-Vargas H, Canto P. In vitro and
28101–28108. bution to testis differentiation in the fetal mouse. molecular modeling analysis of two mutant Desert
247. O’Bryan MK, Takada S, Kennedy CL, Scott G, Harada Development. 1993;117(1):273–281. hedgehog proteins associated with 46,XY gonadal
S, Ray MK, Dai Q, Wilhelm D, de Kretser DM, Eddy 265. Cool J, Carmona FD, Szucsik JC, Capel B. Peritubular dysgenesis. DNA Cell Biol. 2013;32(9):524–530.
EM, Koopman P, Mishina Y. Sox8 is a critical reg- myoid cells are not the migrating population re- 282. Werner R, Merz H, Birnbaum W, Marshall L,
ulator of adult Sertoli cell function and male fertility. quired for testis cord formation in the XY gonad. Schröder T, Reiz B, Kavran JM, Bäumer T, Capetian P,
Dev Biol. 2008;316(2):359–370. Sex Dev. 2008;2(3):128–133. Hiort O. 46,XY gonadal dysgenesis due to a ho-
248. Singh AP, Cummings CA, Mishina Y, Archer TK. 266. Jeanes A, Wilhelm D, Wilson MJ, Bowles J, McClive mozygous mutation in desert hedgehog (DHH)
SOX8 regulates permeability of the blood-testes PJ, Sinclair AH, Koopman P. Evaluation of candidate identified by exome sequencing. J Clin Endocrinol
barrier that affects adult male fertility in the markers for the peritubular myoid cell lineage in the Metab. 2015;100(7):E1022–E1029.
mouse. Biol Reprod. 2013;88(5):133. developing mouse testis. Reproduction. 2005;130(4): 283. Hutson JM. A biphasic model for the hormonal
249. Barrionuevo F, Georg I, Scherthan H, Lécureuil C, 509–516. control of testicular descent. Lancet. 1985;2(8452):
Guillou F, Wegner M, Scherer G. Testis cord dif- 267. Davidoff MS, Breucker H, Holstein AF, Seidl K. 419–421.
ferentiation after the sex determination stage is Cellular architecture of the lamina propria of hu- 284. Hutson JM, Li R, Southwell BR, Newgreen D,
independent of Sox9 but fails in the combined man seminiferous tubules. Cell Tissue Res. 1990; Cousinery M. Regulation of testicular descent.
absence of Sox9 and Sox8. Dev Biol. 2009;327(2): 262(2):253–261. Pediatr Surg Int. 2015;31(4):317–325.
301–312. 268. Tung PS, Skinner MK, Fritz IB. Cooperativity be- 285. Hutson JM, Baskin LS, Risbridger G, Cunha GR. The

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


250. Moalem S, Babul-Hirji R, Stavropolous DJ, Wherrett tween Sertoli cells and peritubular myoid cells in power and perils of animal models with urogenital
D, Bägli DJ, Thomas P, Chitayat D. XX male sex the formation of the basal lamina in the seminif- anomalies: handle with care. J Pediatr Urol. 2014;
reversal with genital abnormalities associated with a erous tubule. Ann N Y Acad Sci. 1984;438(1): 10(4):699–705.
de novo SOX3 gene duplication. Am J Med Genet A. 435–446. 286. Hutson JM, Li R, Vikraman J, Loebenstein M. What
2012;158A(7):1759–1764. 269. Gaytan F, Bellido C, Aguilar E, van Rooijen N. Re- animal models of testicular descent and germ cell
251. Zhao L, Arsenault M, Ng ET, Longmuss E, Chau TC, quirement for testicular macrophages in Leydig cell maturation tell us about the mechanism in
Hartwig S, Koopman P. SOX4 regulates gonad proliferation and differentiation during prepubertal humans. Eur J Pediatr Surg. 2016;26(5):390–398.
morphogenesis and promotes male germ cell dif- development in rats. J Reprod Fertil. 1994;102(2): 287. Nef S, Parada LF. Cryptorchidism in mice mutant for
ferentiation in mice. Dev Biol. 2017;423(1):46–56. 393–399. Insl3. Nat Genet. 1999;22(3):295–299.
252. Nel-Themaat L, Vadakkan TJ, Wang Y, Dickinson 270. Hutson JC. Development of cytoplasmic digitations 288. Zimmermann S, Steding G, Emmen JM, Brinkmann
ME, Akiyama H, Behringer RR. Morphometric between Leydig cells and testicular macrophages of AO, Nayernia K, Holstein AF, Engel W, Adham IM.
analysis of testis cord formation in Sox9-EGFP mice. the rat. Cell Tissue Res. 1992;267(2):385–389. Targeted disruption of the Insl3 gene causes bi-
Dev Dyn. 2009;238(5):1100–1110. 271. DeFalco T, Potter SJ, Williams AV, Waller B, Kan MJ, lateral cryptorchidism. Mol Endocrinol. 1999;13(5):
253. Miles DC, Wakeling SI, Stringer JM, van den Bergen Capel B. Macrophages contribute to the sper-
681–691.
JA, Wilhelm D, Sinclair AH, Western PS. Signaling matogonial niche in the adult testis. Cell Reports.
289. Gorlov IP, Kamat A, Bogatcheva NV, Jones E, Lamb
through the TGF beta-activin receptors ALK4/5/7 2015;12(7):1107–1119.
DJ, Truong A, Bishop CE, McElreavey K, Agoulnik AI.
regulates testis formation and male germ cell de- 272. Mossadegh-Keller N, Gentek R, Gimenez G, Bigot S,
Mutations of the GREAT gene cause cryptorchi-
velopment. PLoS One. 2013;8(1):e54606. Mailfert S, Sieweke MH. Developmental origin and
dism. Hum Mol Genet. 2002;11(19):2309–2318.
254. McCoshen JA. In vivo sex differentiation of con- maintenance of distinct testicular macrophage
290. Shono T, Ramm-Anderson S, Hutson JM. Trans-
geneic germinal cell aplastic gonads. Am J Obstet populations. J Exp Med. 2017;214(10):2829–2841.
abdominal testicular descent is really ovarian ascent.
Gynecol. 1982;142(1):83–88. 273. Bitgood MJ, Shen L, McMahon AP. Sertoli cell
J Urol. 1994;152(2 Pt 2):781–784.
255. McCoshen JA. Quantitation of sex chromosomal signaling by Desert hedgehog regulates the male
291. Kaftanovskaya EM, Feng S, Huang Z, Tan Y, Barbara
influence(s) on the somatic growth of fetal gonads germline. Curr Biol. 1996;6(3):298–304.
AM, Kaur S, Truong A, Gorlov IP, Agoulnik AI.
in vivo. Am J Obstet Gynecol. 1983;145(4):469–473. 274. Pierucci-Alves F, Clark AM, Russell LD. A de-
Suppression of insulin-like3 receptor reveals the role
256. Rios-Rojas C, Spiller C, Bowles J, Koopman P. Germ velopmental study of the Desert hedgehog-null
of b-catenin and Notch signaling in gubernaculum
cells influence cord formation and Leydig cell gene mouse testis. Biol Reprod. 2001;65(5):1392–1402.
development. Mol Endocrinol. 2011;25(1):170–183.
expression during mouse testis development. Dev 275. Yao HH, Capel B. Disruption of testis cords by
292. Huang Z, Rivas B, Agoulnik AI. Insulin-like 3 signaling
Dyn. 2016;245(4):433–444. cyclopamine or forskolin reveals independent cel-
257. Martineau J, Nordqvist K, Tilmann C, Lovell-Badge R, lular pathways in testis organogenesis. Dev Biol. is important for testicular descent but dispensable
Capel B. Male-specific cell migration into the de- 2002;246(2):356–365. for spermatogenesis and germ cell survival in adult
veloping gonad. Curr Biol. 1997;7(12):958–968. 276. Barsoum IB, Bingham NC, Parker KL, Jorgensen JS, mice. Biol Reprod. 2012;87(6):143.
258. Tilmann C, Capel B. Mesonephric cell migration Yao HH. Activation of the Hedgehog pathway in 293. Barteczko KJ, Jacob MI. The testicular descent in
induces testis cord formation and Sertoli cell dif- the mouse fetal ovary leads to ectopic appearance human. Origin, development and fate of the
ferentiation in the mammalian gonad. Development. of fetal Leydig cells and female pseudohermaph- gubernaculum Hunteri, processus vaginalis peri-
1999;126(13):2883–2890. roditism. Dev Biol. 2009;329(1):96–103. tonei, and gonadal ligaments. Adv Anat Embryol Cell
259. Smith CA, McClive PJ, Hudson Q, Sinclair AH. Male- 277. Park SY, Tong M, Jameson JL. Distinct roles for Biol. 2000;156:III–X, 1–98.
specific cell migration into the developing gonad steroidogenic factor 1 and desert hedgehog pathways 294. Bouzada J, Vázquez T, Duran M, Delmas V, Larkin T,
is a conserved process involving PDGF signalling. in fetal and adult Leydig cell development. Endo- Cuesta MA, SañUDO J. New insights into the
Dev Biol. 2005;284(2):337–350. crinology. 2007;148(8):3704–3710. morphogenesis of the gubernaculum testis and the
260. Coveney D, Cool J, Oliver T, Capel B. Four- 278. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, inguinal canal. Clin Anat. 2017;30(5):599–607.
dimensional analysis of vascularization during pri- Mitsuya T, Osame M. A novel mutation of desert 295. Hadziselimovic F. On the descent of the epididymo-
mary development of an organ, the gonad. Proc hedgehog in a patient with 46,XY partial gonadal testicular unit, cryptorchidism, and prevention of
Natl Acad Sci USA. 2008;105(20):7212–7217. dysgenesis accompanied by minifascicular neu- infertility. Basic Clin Androl. 2017;27(1):21.
261. Bott RC, McFee RM, Clopton DT, Toombs C, Cupp ropathy. Am J Hum Genet. 2000;67(5):1302–1305. 296. Anand-Ivell R, Ivell R, Driscoll D, Manson J. Insulin-
AS. Vascular endothelial growth factor and kinase 279. Canto P, Söderlund D, Reyes E, Méndez JP. Muta- like factor 3 levels in amniotic fluid of human male
domain region receptor are involved in both tions in the Desert hedgehog (DHH) gene in patients fetuses. Hum Reprod. 2008;23(5):1180–1186.
seminiferous cord formation and vascular devel- with 46,XY complete pure gonadal dysgenesis. J Clin 297. Bay K, Cohen AS, Jørgensen FS, Jørgensen C, Lind
opment during testis morphogenesis in the rat. Biol Endocrinol Metab. 2004;89(9):4480–4483. AM, Skakkebaek NE, Andersson AM. Insulin-like
Reprod. 2006;75(1):56–67. 280. Das DK, Sanghavi D, Gawde H, Idicula-Thomas S, factor 3 levels in second-trimester amniotic fluid.
262. Cool J, DeFalco TJ, Capel B. Vascular-mesenchymal Vasudevan L. Novel homozygous mutations in J Clin Endocrinol Metab. 2008;93(10):4048–4051.
cross-talk through Vegf and Pdgf drives organ Desert Hedgehog gene in patients with 46,XY 298. Bay K, Virtanen HE, Hartung S, Ivell R, Main KM,
patterning. Proc Natl Acad Sci USA. 2011;108(1): complete gonadal dysgenesis and prediction of its Skakkebaek NE, Andersson AM, Toppari J; Nordic
167–172. structural and functional implications by compu- Cryptorchidism Study Group. Insulin-like factor 3
263. Combes AN, Wilhelm D, Davidson T, Dejana E, tational methods. Eur J Med Genet. 2011;54(6): levels in cord blood and serum from children: ef-
Harley V, Sinclair A, Koopman P. Endothelial cell e529–e534. fects of age, postnatal hypothalamic-pituitary-
migration directs testis cord formation. Dev Biol. 281. Castro JJ, Méndez JP, Coral-Vázquez RM, Soriano- gonadal axis activation, and cryptorchidism. J Clin
2009;326(1):112–120. Ursúa MA, Damian-Matsumura P, Benı́t ez- Endocrinol Metab. 2007;92(10):4020–4027.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 899


REVIEW

299. Fénichel P, Lahlou N, Coquillard P, Panaı̈a-Ferrari P, 316. Yuan FP, Li X, Lin J, Schwabe C, Büllesbach EE, Rao development in the XY gonad. Genes Dev. 2003;
Wagner-Mahler K, Brucker-Davis F. Cord blood CV, Lei ZM. The role of RXFP2 in mediating 17(6):800–810.
insulin-like peptide 3 (INSL3) but not testosterone androgen-induced inguinoscrotal testis descent in 332. O’Hara L, McInnes K, Simitsidellis I, Morgan S,
is reduced in idiopathic cryptorchidism. Clin LH receptor knockout mice. Reproduction. 2010; Atanassova N, Slowikowska-Hilczer J, Kula K,
Endocrinol (Oxf). 2015;82(2):242–247. 139(4):759–769. Szarras-Czapnik M, Milne L, Mitchell RT, Smith LB.
300. Mamoulakis C, Georgiou I, Dimitriadis F, Tsounapi P, 317. Virtanen HE, Toppari J. Epidemiology and patho- Autocrine androgen action is essential for Leydig
Giannakis I, Chatzikyriakidou A, Antypas S, Sofras F, genesis of cryptorchidism. Hum Reprod Update. cell maturation and function, and protects against
Takenaka A, Sofikitis N. Genetic analysis of the 2008;14(1):49–58. late-onset Leydig cell apoptosis in both mice and
human insulin-like 3 gene: absence of mutations 318. Cendron M, Huff DS, Keating MA, Snyder HM III, men. FASEB J. 2015;29(3):894–910.
in a Greek paediatric cohort with testicular mal- Duckett JW. Anatomical, morphological and vol- 333. Baker PJ, Sha JH, O’Shaughnessy PJ. Localisation and
descent. Andrologia. 2014;46(9):986–996. umetric analysis: a review of 759 cases of testicular regulation of 17b-hydroxysteroid dehydrogenase
301. Virtanen HE, Toppari J. Embryology and physiology maldescent. J Urol. 1993;149(3):570–573. type 3 mRNA during development in the mouse
of testicular development and descent. Pediatr 319. Raivio T, Toppari J, Kaleva M, Virtanen H, Haavisto testis. Mol Cell Endocrinol. 1997;133(2):127–133.
Endocrinol Rev. 2014;11(Suppl 2):206–213. AM, Dunkel L, Jänne OA. Serum androgen bio- 334. Shima Y, Miyabayashi K, Haraguchi S, Arakawa T,
302. Hutson JM, Lopez-Marambio FA. The possible role activity in cryptorchid and noncryptorchid boys Otake H, Baba T, Matsuzaki S, Shishido Y, Akiyama
of AMH in shortening the gubernacular cord in during the postnatal reproductive hormone surge. H, Tachibana T, Tsutsui K, Morohashi K. Contri-
testicular descent: a reappraisal of the evidence. J Clin Endocrinol Metab. 2003;88(6):2597–2599. bution of Leydig and Sertoli cells to testosterone

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


J Pediatr Surg. 2017;52(10):1656–1660. 320. Suomi AM, Main KM, Kaleva M, Schmidt IM, production in mouse fetal testes. Mol Endocrinol.
303. Behringer RR, Finegold MJ, Cate RL. Müllerian- Chellakooty M, Virtanen HE, Boisen KA, Damgaard 2013;27(1):63–73.
inhibiting substance function during mammalian IN, Kai CM, Skakkebaek NE, Toppari J. Hormonal 335. Huhtaniemi I, Alevizaki M. Gonadotrophin re-
sexual development. Cell. 1994;79(3):415–425. changes in 3-month-old cryptorchid boys. J Clin sistance. Best Pract Res Clin Endocrinol Metab. 2006;
304. Nation TR, Buraundi S, Farmer PJ, Balic A, Newgreen Endocrinol Metab. 2006;91(3):953–958. 20(4):561–576.
D, Southwell BR, Hutson JM. Development of the 321. Kuiri-Hänninen T, Seuri R, Tyrväinen E, Turpeinen U, 336. Känsäkoski J, Jääskeläinen J, Jääskeläinen T,
gubernaculum during testicular descent in the rat. Hämäläinen E, Stenman UH, Dunkel L, Sankilampi Tommiska J, Saarinen L, Lehtonen R, Hautaniemi S,
Anat Rec (Hoboken). 2011;294(7):1249–1260. U. Increased activity of the hypothalamic-pituitary- Frilander MJ, Palvimo JJ, Toppari J, Raivio T. Com-
305. Na AF, Harnaen EJ, Farmer PJ, Sourial M, Southwell testicular axis in infancy results in increased an- plete androgen insensitivity syndrome caused by a
BR, Hutson JM. Cell membrane and mitotic markers drogen action in premature boys. J Clin Endocrinol deep intronic pseudoexon-activating mutation in
show that the neonatal rat gubernaculum grows Metab. 2011;96(1):98–105. the androgen receptor gene. Sci Rep. 2016;6(1):
in a similar way to an embryonic limb bud. J Pediatr 322. Koskenniemi JJ, Virtanen HE, Wohlfahrt-Veje C, 32819.
Surg. 2007;42(9):1566–1573. Löyttyniemi E, Skakkebaek NE, Juul A, Andersson 337. Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T,
306. Heyns CF. The gubernaculum during testicular AM, Main KM, Toppari J. Postnatal changes in Sekido R, Kress J, Treier AC, Klugmann C, Klasen C,
descent in the human fetus. J Anat. 1987;153: testicular position are associated with IGF-I and Holter NI, Riethmacher D, Schütz G, Cooney AJ,
93–112. function of Sertoli and Leydig cells. J Clin Endocrinol Lovell-Badge R, Treier M. Somatic sex reprogram-
307. Achiron R, Pinhas-Hamiel O, Zalel Y, Rotstein Z, Metab. 2018;103(4):1429–1437. ming of adult ovaries to testes by FOXL2 ablation.
Lipitz S. Development of fetal male gender: prenatal 323. Moorthy AV, Chesney RW, Lubinsky M. Chronic Cell. 2009;139(6):1130–1142.
sonographic measurement of the scrotum and renal failure and XY gonadal dysgenesis: “Frasier” 338. Moniot B, Berta P, Scherer G, Südbeck P, Poulat F.
evaluation of testicular descent. Ultrasound Obstet syndrome—a commentary on reported cases. Am J Male specific expression suggests role of DMRT1 in
Gynecol. 1998;11(4):242–245. Med Genet Suppl. 1987;28(Suppl 3):297–302. human sex determination. Mech Dev. 2000;91(1–2):
308. Sampaio FJ, Favorito LA. Analysis of testicular mi- 324. Tachibana M. Epigenetic regulation of mammalian 323–325.
gration during the fetal period in humans. J Urol. sex determination. J Med Invest. 2015;62(1–2): 339. Jørgensen A, Nielsen JE, Blomberg Jensen M, Græm
1998;159(2):540–542. 19–23. N, Rajpert-De Meyts E. Analysis of meiosis regulators
309. Rotondi M, Valenzano F, Bilancioni E, Spanò G, 325. Knarston I, Ayers K, Sinclair A. Molecular mecha- in human gonads: a sexually dimorphic spatio-
Rotondi M, Giorlandino C. Prenatal measurement nisms associated with 46,XX disorders of sex de- temporal expression pattern suggests involvement
of testicular diameter by ultrasonography: devel- velopment. Clin Sci (Lond). 2016;130(6):421–432. of DMRT1 in meiotic entry. Mol Hum Reprod. 2012;
opment of fetal male gender and evaluation of 326. Wirth J, Wagner T, Meyer J, Pfeiffer RA, Tietze HU, 18(11):523–534.
testicular descent. Prenat Diagn. 2001;21(2): Schempp W, Scherer G. Translocation breakpoints 340. De Grandi A, Calvari V, Bertini V, Bulfone A, Peverali
112–115. in three patients with campomelic dysplasia and G, Camerino G, Borsani G, Guioli S. The expression
310. Hutson JM. Testicular feminization: a model for autosomal sex reversal map more than 130 kb from pattern of a mouse doublesex-related gene is
testicular descent in mice and men. J Pediatr Surg. SOX9. Hum Genet. 1996;97(2):186–193. consistent with a role in gonadal differentiation.
1986;21(3):195–198. 327. Falah N, Posey JE, Thorson W, Benke P, Tekin M, Mech Dev. 2000;90(2):323–326.
311. Barthold JS, Kumasi-Rivers K, Upadhyay J, Shekarriz Tarshish B, Lupski JR, Harel T. 22q11.2q13 duplica- 341. Raymond CS, Shamu CE, Shen MM, Seifert KJ,
B, Imperato-Mcginley J. Testicular position in the tion including SOX10 causes sex-reversal and Hirsch B, Hodgkin J, Zarkower D. Evidence for
androgen insensitivity syndrome: implications for peripheral demyelinating neuropathy, central evolutionary conservation of sex-determining
the role of androgens in testicular descent. J Urol. dysmyelinating leukodystrophy, Waardenburg genes. Nature. 1998;391(6668):691–695.
2000;164(2):497–501. syndrome, and Hirschsprung disease. Am J Med 342. Veitia R, Nunes M, Brauner R, Doco-Fenzy M,
312. Ferlin A, Zuccarello D, Zuccarello B, Chirico MR, Genet A. 2017;173(4):1066–1070. Joanny-Flinois O, Jaubert F, Lortat-Jacob S, Fellous
Zanon GF, Foresta C. Genetic alterations associated 328. Jørgensen A, Rajpert-De Meyts E. Regulation of M, McElreavey K. Deletions of distal 9p associated
with cryptorchidism. JAMA. 2008;300(19):2271–2276. meiotic entry and gonadal sex differentiation in the with 46,XY male to female sex reversal: definition of
313. Spencer JR, Torrado T, Sanchez RS, Vaughan ED Jr, human: normal and disrupted signaling. Biomol the breakpoints at 9p23.3–p24.1. Genomics. 1997;
Imperato-McGinley J. Effects of flutamide and Concepts. 2014;5(4):331–341. 41(2):271–274.
finasteride on rat testicular descent. Endocrinology. 329. Sirisena ND, McElreavey K, Bashamboo A, de Silva 343. Tannour-Louet M, Han S, Corbett ST, Louet JF,
1991;129(2):741–748. KS, Jayasekara RW, Dissanayake VH. A child with a Yatsenko S, Meyers L, Shaw CA, Kang SH, Cheung
314. Welsh M, Saunders PT, Fisken M, Scott HM, novel de novo mutation in the aristaless domain of SW, Lamb DJ. Identification of de novo copy
Hutchison GR, Smith LB, Sharpe RM. Identification the aristaless-related homeobox (ARX) gene pre- number variants associated with human disorders
in rats of a programming window for reproductive senting with ambiguous genitalia and psychomotor of sexual development. PLoS One. 2010;5(10):
tract masculinization, disruption of which leads to delay. Sex Dev. 2014;8(4):156–159. e15392.
hypospadias and cryptorchidism. J Clin Invest. 2008; 330. Ruiz-Arana IL, Hübner A, Cetingdag C, Krude H, 344. Raymond CS, Murphy MW, O’Sullivan MG,
118(4):1479–1490. Grüters A, Fukami M, Biebermann H, Köhler B. A Bardwell VJ, Zarkower D. Dmrt1, a gene related to
315. Cousinery MC, Li R, Vannitamby A, Vikraman J, novel hemizygous mutation of MAMLD1 in a pa- worm and fly sexual regulators, is required for
Southwell BR, Hutson JM. Neurotrophin signaling tient with 46,XY complete gonadal dysgenesis. Sex mammalian testis differentiation. Genes Dev. 2000;
in a genitofemoral nerve target organ during tes- Dev. 2015;9(2):80–85. 14(20):2587–2595.
ticular descent in mice. J Pediatr Surg. 2016;51(8): 331. Brennan J, Tilmann C, Capel B. Pdgfr-a mediates 345. Fahrioglu U, Murphy MW, Zarkower D, Bardwell VJ.
1321–1326. testis cord organization and fetal Leydig cell mRNA expression analysis and the molecular basis

900 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

of neonatal testis defects in Dmrt1 mutant mice. Sex 361. Molyneaux K, Wylie C. Primordial germ cell mi- 378. Snyder EM, Small C, Griswold MD. Retinoic acid
Dev. 2007;1(1):42–58. gration. Int J Dev Biol. 2004;48(5–6):537–544. availability drives the asynchronous initiation of
346. Kim S, Bardwell VJ, Zarkower D. Cell type- 362. Culty M. Gonocytes, from the fifties to the present: spermatogonial differentiation in the mouse. Biol
autonomous and non-autonomous requirements is there a reason to change the name? Biol Reprod. Reprod. 2010;83(5):783–790.
for Dmrt1 in postnatal testis differentiation. Dev Biol. 2013;89(2):46. 379. Snyder EM, Davis JC, Zhou Q, Evanoff R, Griswold
2007;307(2):314–327. 363. McCarrey JR. Toward a more precise and in- MD. Exposure to retinoic acid in the neonatal but
347. De Baere E, Dixon MJ, Small KW, Jabs EW, Leroy BP, formative nomenclature describing fetal and neo- not adult mouse results in synchronous sper-
Devriendt K, Gillerot Y, Mortier G, Meire F, Van natal male germ cells in rodents. Biol Reprod. 2013; matogenesis. Biol Reprod. 2011;84(5):886–893.
Maldergem L, Courtens W, Hjalgrim H, Huang S, 89(2):47. 380. Koubova J, Menke DB, Zhou Q, Capel B, Griswold
Liebaers I, Van Regemorter N, Touraine P, 364. Hilscher B, Hilscher W, Bülthoff-Ohnolz B, Krämer MD, Page DC. Retinoic acid regulates sex-specific
Praphanphoj V, Verloes A, Udar N, Yellore V, U, Birke A, Pelzer H, Gauss G. Kinetics of game- timing of meiotic initiation in mice. Proc Natl Acad
Chalukya M, Yelchits S, De Paepe A, Kuttenn F, togenesis. I. Comparative histological and autora- Sci USA. 2006;103(8):2474–2479.
Fellous M, Veitia R, Messiaen L. Spectrum of FOXL2 diographic studies of oocytes and transitional 381. Bowles J, Knight D, Smith C, Wilhelm D, Richman J,
gene mutations in blepharophimosis-ptosis-epicanthus prospermatogonia during oogenesis and pre- Mamiya S, Yashiro K, Chawengsaksophak K, Wilson
inversus (BPES) families demonstrates a genotype– spermatogenesis. Cell Tissue Res. 1974;154(4): MJ, Rossant J, Hamada H, Koopman P. Retinoid
phenotype correlation. Hum Mol Genet. 2001;10(15): 443–470. signaling determines germ cell fate in mice. Science.
1591–1600. 365. Nagano R, Tabata S, Nakanishi Y, Ohsako S, 2006;312(5773):596–600.

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


348. Pailhoux E, Vigier B, Chaffaux S, Servel N, Taourit S, Kurohmaru M, Hayashi Y. Reproliferation and re- 382. Barrios F, Filipponi D, Pellegrini M, Paronetto MP, Di
Furet JP, Fellous M, Grosclaude F, Cribiu EP, Cotinot location of mouse male germ cells (gonocytes) Siena S, Geremia R, Rossi P, De Felici M, Jannini EA,
C, Vaiman D. A 11.7-kb deletion triggers in- during prespermatogenesis. Anat Rec. 2000;258(2): Dolci S. Opposing effects of retinoic acid and FGF9
tersexuality and polledness in goats. Nat Genet. 210–220. on Nanos2 expression and meiotic entry of mouse
2001;29(4):453–458. 366. Western PS, Miles DC, van den Bergen JA, Burton M, germ cells. J Cell Sci. 2010;123(Pt 6):871–880.
349. Fokstuen S, Antonarakis SE, Blouin JL. FOXL2-mu- Sinclair AH. Dynamic regulation of mitotic arrest in 383. Bowles J, Feng CW, Spiller C, Davidson TL, Jackson A,
tations in blepharophimosis-ptosis-epicanthus fetal male germ cells. Stem Cells. 2008;26(2):339–347. Koopman P. FGF9 suppresses meiosis and promotes
inversus syndrome (BPES); challenges for genetic 367. Pui HP, Saga Y. Gonocytes-to-spermatogonia male germ cell fate in mice. Dev Cell. 2010;19(3):
counseling in female patients. Am J Med Genet A. transition initiates prior to birth in murine testes 440–449.
2003;117A(2):143–146. and it requires FGF signaling. Mech Dev. 2017;144(Pt 384. Spiller CM, Feng CW, Jackson A, Gillis AJ, Rolland
350. Uhlenhaut NH, Treier M. Foxl2 function in ovarian B):125–139. AD, Looijenga LH, Koopman P, Bowles J. Endoge-
development. Mol Genet Metab. 2006;88(3): 368. Zhou Q, Li Y, Nie R, Friel P, Mitchell D, Evanoff RM, nous Nodal signaling regulates germ cell potency
Pouchnik D, Banasik B, McCarrey JR, Small C, during mammalian testis development. Develop-
225–234.
Griswold MD. Expression of stimulated by retinoic ment. 2012;139(22):4123–4132.
351. Boulanger L, Pannetier M, Gall L, Allais-Bonnet A,
acid gene 8 (Stra8) and maturation of murine 385. Kashimada K, Svingen T, Feng CW, Pelosi E, Bagheri-
Elzaiat M, Le Bourhis D, Daniel N, Richard C, Cotinot
gonocytes and spermatogonia induced by retinoic Fam S, Harley VR, Schlessinger D, Bowles J, Koopman
C, Ghyselinck NB, Pailhoux E. FOXL2 is a female sex-
acid in vitro. Biol Reprod. 2008;78(3):537–545. P. Antagonistic regulation of Cyp26b1 by tran-
determining gene in the goat. Curr Biol. 2014;24(4):
369. Tam PP, Snow MH. Proliferation and migration of scription factors SOX9/SF1 and FOXL2 during go-
404–408.
primordial germ cells during compensatory growth nadal development in mice. FASEB J. 2011;25(10):
352. Minkina A, Matson CK, Lindeman RE, Ghyselinck
in mouse embryos. J Embryol Exp Morphol. 1981;64: 3561–3569.
NB, Bardwell VJ, Zarkower D. DMRT1 protects male
133–147. 386. Suzuki A, Saga Y. Nanos2 suppresses meiosis and
gonadal cells from retinoid-dependent sexual
370. Vergouwen RP, Jacobs SG, Huiskamp R, Davids JA, promotes male germ cell differentiation. Genes Dev.
transdifferentiation. Dev Cell. 2014;29(5):511–520.
de Rooij DG. Proliferative activity of gonocytes, 2008;22(4):430–435.
353. De Gendt K, Verhoeven G, Amieux PS, Wilkinson
Sertoli cells and interstitial cells during testicular 387. Souquet B, Tourpin S, Messiaen S, Moison D, Habert
MF. Genome-wide identification of AR-regulated
development in mice. J Reprod Fertil. 1991;93(1): R, Livera G. Nodal signaling regulates the entry into
genes translated in Sertoli cells in vivo using the 233–243. meiosis in fetal germ cells. Endocrinology. 2012;
RiboTag approach. Mol Endocrinol. 2014;28(4): 371. Heaney JD, Anderson EL, Michelson MV, Zechel JL, 153(5):2466–2473.
575–591. Conrad PA, Page DC, Nadeau JH. Germ cell plu- 388. Richards AJ, Enders GC, Resnick JL. Activin and
354. Zhao L, Svingen T, Ng ET, Koopman P. Female-to- ripotency, premature differentiation and suscepti- TGFb limit murine primordial germ cell pro-
male sex reversal in mice caused by transgenic bility to testicular teratomas in mice. Development. liferation. Dev Biol. 1999;207(2):470–475.
overexpression of Dmrt1. Development. 2015;142(6): 2012;139(9):1577–1586. 389. Godin I, Wylie CC. TGF beta 1 inhibits proliferation
1083–1088. 372. Suzuki A, Tsuda M, Saga Y. Functional redundancy and has a chemotropic effect on mouse primordial
355. Lindeman RE, Gearhart MD, Minkina A, Krentz AD, among Nanos proteins and a distinct role of germ cells in culture. Development. 1991;113(4):
Bardwell VJ, Zarkower D. Sexual cell-fate reprog- Nanos2 during male germ cell development. De- 1451–1457.
ramming in the ovary by DMRT1. Curr Biol. 2015; velopment. 2007;134(1):77–83. 390. Kaipia A, Toppari J, Huhtaniemi I, Paranko J. Sex
25(6):764–771. 373. Pui HP, Saga Y. NANOS2 acts as an intrinsic reg- difference in the action of activin-A on cell pro-
356. Barrionuevo FJ, Hurtado A, Kim GJ, Real FM, Bakkali ulator of gonocytes-to-spermatogonia transition in liferation of differentiating rat gonad. Endocrinology.
M, Kopp JL, Sander M, Scherer G, Burgos M, Jiménez the murine testes. Mech Dev. 2018;149:27–40. 1994;134(5):2165–2170.
R. Sox9 and Sox8 protect the adult testis from male- 374. Roosen-Runge EC, Leik J. Gonocyte degeneration in 391. Moreno SG, Attali M, Allemand I, Messiaen S,
to-female genetic reprogramming and complete the postnatal male rat. Am J Anat. 1968;122(2): Fouchet P, Coffigny H, Romeo PH, Habert R. TGFb
degeneration. eLife. 2016;5:e15635. 275–299. signaling in male germ cells regulates gonocyte
357. Jameson SA, Lin YT, Capel B. Testis development 375. Beaumont HM, Mandl AM. A quantitative study of quiescence and fertility in mice. Dev Biol. 2010;
requires the repression of Wnt4 by Fgf signaling. Dev primordial germ cells in the male rat. J Embryol Exp 342(1):74–84.
Biol. 2012;370(1):24–32. Morphol. 1963;11:715–740. 392. Mendis SH, Meachem SJ, Sarraj MA, Loveland KL.
358. Maatouk DM, DiNapoli L, Alvers A, Parker KL, 376. Yoshida S, Sukeno M, Nakagawa T, Ohbo K, Activin A balances Sertoli and germ cell pro-
Taketo MM, Capel B. Stabilization of b-catenin in Nagamatsu G, Suda T, Nabeshima Y. The first round liferation in the fetal mouse testis. Biol Reprod. 2011;
XY gonads causes male-to-female sex-reversal. Hum of mouse spermatogenesis is a distinctive program 84(2):379–391.
Mol Genet. 2008;17(19):2949–2955. that lacks the self-renewing spermatogonia stage. 393. Spiller CM, Wilhelm D, Koopman P. Retinoblastoma
359. Chang H, Gao F, Guillou F, Taketo MM, Huff V, Development. 2006;133(8):1495–1505. 1 protein modulates XY germ cell entry into G1/G0
Behringer RR. Wt1 negatively regulates b-catenin 377. Tokue M, Ikami K, Mizuno S, Takagi C, Miyagi A, arrest during fetal development in mice. Biol Reprod.
signaling during testis development. Development. Takada R, Noda C, Kitadate Y, Hara K, Mizuguchi H, 2010;82(2):433–443.
2008;135(10):1875–1885. Sato T, Taketo MM, Sugiyama F, Ogawa T, 394. Rossitto M, Ujjan S, Poulat F, Boizet-Bonhoure B.
360. Jordan BK, Mohammed M, Ching ST, Délot E, Chen Kobayashi S, Ueno N, Takahashi S, Takada S, Multiple roles of the prostaglandin D2 signaling
XN, Dewing P, Swain A, Rao PN, Elejalde BR, Vilain E. Yoshida S. SHISA6 confers resistance to pathway in reproduction. Reproduction. 2015;
Up-regulation of WNT-4 signaling and dosage- differentiation-promoting Wnt/b-catenin signaling 149(1):R49–R58.
sensitive sex reversal in humans. Am J Hum in mouse spermatogenic stem cells. Stem Cell Re- 395. Moniot B, Ujjan S, Champagne J, Hirai H, Aritake K,
Genet. 2001;68(5):1102–1109. ports. 2017;8(3):561–575. Nagata K, Dubois E, Nidelet S, Nakamura M, Urade

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 901


REVIEW

Y, Poulat F, Boizet-Bonhoure B. Prostaglandin D2 SALL4-dependent silencing of distinct tumor sup- identification of signaling mechanisms involved.
acts through the Dp2 receptor to influence male pressor genes. Stem Cell Reports. 2017;9(3):956–971. Endocrinology. 1997;138(3):1289–1298.
germ cell differentiation in the foetal mouse testis. 411. Kasowitz SD, Luo M, Ma J, Leu NA, Wang PJ. 427. Thuillier R, Mazer M, Manku G, Boisvert A, Wang Y,
Development. 2014;141(18):3561–3571. Embryonic lethality and defective male germ cell Culty M. Interdependence of platelet-derived
396. Kimura T, Suzuki A, Fujita Y, Yomogida K, Lomeli H, development in mice lacking UTF1. Sci Rep. 2017; growth factor and estrogen-signaling pathways in
Asada N, Ikeuchi M, Nagy A, Mak TW, Nakano T. 7(1):17259. inducing neonatal rat testicular gonocytes pro-
Conditional loss of PTEN leads to testicular tera- 412. Tian-Zhong M, Bi C, Ying Z, Xia J, Cai-Ling P, Yun- liferation. Biol Reprod. 2010;82(5):825–836.
toma and enhances embryonic germ cell pro- Shan Z, Mei-Wen H, Yan-Ru N. Critical role of Emx2 428. Manku G, Culty M. Mammalian gonocyte and
duction. Development. 2003;130(8):1691–1700. in the pluripotency—differentiation transition in spermatogonia differentiation: recent advances and
397. Krentz AD, Murphy MW, Kim S, Cook MS, Capel B, male gonocytes via regulation of FGF9/NODAL remaining challenges. Reproduction. 2015;149(3):
Zhu R, Matin A, Sarver AL, Parker KL, Griswold MD, pathway. Reproduction. 2016;151(6):673–681. R139–R157.
Looijenga LH, Bardwell VJ, Zarkower D. The DM 413. Oosterhuis JW, Looijenga LH. Testicular germ-cell 429. Xu J, Wan P, Wang M, Zhang J, Gao X, Hu B, Han J,
domain protein DMRT1 is a dose-sensitive regu- tumours in a broader perspective. Nat Rev Cancer. Chen L, Sun K, Wu J, Wu X, Huang X, Chen J. AIP1-
lator of fetal germ cell proliferation and pluripo- 2005;5(3):210–222. mediated actin disassembly is required for postnatal
tency. Proc Natl Acad Sci USA. 2009;106(52): 414. Hussain SA, Ma YT, Palmer DH, Hutton P, Cullen germ cell migration and spermatogonial stem cell
22323–22328. MH. Biology of testicular germ cell tumors. Expert niche establishment. Cell Death Dis. 2015;6(7):e1818.
398. Cook MS, Munger SC, Nadeau JH, Capel B. Regu- Rev Anticancer Ther. 2008;8(10):1659–1673. 430. Orwig KE, Ryu BY, Avarbock MR, Brinster RL. Male

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


lation of male germ cell cycle arrest and differen- 415. Sonne SB, Almstrup K, Dalgaard M, Juncker AS, germ-line stem cell potential is predicted by
tiation by DND1 is modulated by genetic Edsgard D, Ruban L, Harrison NJ, Schwager C, morphology of cells in neonatal rat testes. Proc Natl
background. Development. 2011;138(1):23–32. Abdollahi A, Huber PE, Brunak S, Gjerdrum LM, Acad Sci USA. 2002;99(18):11706–11711.
399. Kedde M, Strasser MJ, Boldajipour B, Oude Vrielink Moore HD, Andrews PW, Skakkebaek NE, Rajpert- 431. Tres LL, Kierszenbaum AL. The ADAM-integrin-
JA, Slanchev K, le Sage C, Nagel R, Voorhoeve PM, De Meyts E, Leffers H. Analysis of gene expression tetraspanin complex in fetal and postnatal testic-
van Duijse J, Ørom UA, Lund AH, Perrakis A, Raz E, profiles of microdissected cell populations indicates ular cords. Birth Defects Res C Embryo Today. 2005;
Agami R. RNA-binding protein Dnd1 inhibits that testicular carcinoma in situ is an arrested 75(2):130–141.
microRNA access to target mRNA. Cell. 2007;131(7): gonocyte. Cancer Res. 2009;69(12):5241–5250. 432. Song HW, Bettegowda A, Lake BB, Zhao AH,
1273–1286. 416. Skakkebaek NE, Berthelsen JG, Giwercman A, Müller Skarbrevik D, Babajanian E, Sukhwani M, Shum EY,
400. Gross-Thebing T, Yigit S, Pfeiffer J, Reichman-Fried J. Carcinoma-in-situ of the testis: possible origin Phan MH, Plank TM, Richardson ME, Ramaiah M,
M, Bandemer J, Ruckert C, Rathmer C, Goudarzi M, from gonocytes and precursor of all types of germ Sridhar V, de Rooij DG, Orwig KE, Zhang K,
Stehling M, Tarbashevich K, Seggewiss J, Raz E. The cell tumours except spermatocytoma. Int J Androl. Wilkinson MF. The homeobox transcription factor
1987;10(1):19–28. RHOX10 drives mouse spermatogonial stem cell
vertebrate protein dead end maintains primordial
417. Zhou Q, Nie R, Li Y, Friel P, Mitchell D, Hess RA, establishment. Cell Reports. 2016;17(1):149–164.
germ cell fate by inhibiting somatic differentiation.
Small C, Griswold MD. Expression of stimulated by 433. Garcia TX, DeFalco T, Capel B, Hofmann MC.
Dev Cell. 2017;43(6):704–715.e5.
retinoic acid gene 8 (Stra8) in spermatogenic cells Constitutive activation of NOTCH1 signaling in
401. Suzuki A, Niimi Y, Shinmyozu K, Zhou Z, Kiso M,
induced by retinoic acid: an in vivo study in vitamin Sertoli cells causes gonocyte exit from quiescence.
Saga Y. Dead end1 is an essential partner of
A-sufficient postnatal murine testes. Biol Reprod. Dev Biol. 2013;377(1):188–201.
NANOS2 for selective binding of target RNAs in
2008;79(1):35–42. 434. Raju P, Nyamsuren G, Elkenani M, Kata A, Tsagaan E,
male germ cell development. EMBO Rep. 2016;17(1):
418. Chassot AA, Le Rolle M, Jourden M, Taketo MM, Engel W, Adham IM. Pelota mediates gonocyte
37–46.
Ghyselinck NB, Chaboissier MC. Constitutive WNT/ maturation and maintenance of spermatogonial
402. Yamaji M, Jishage M, Meyer C, Suryawanshi H, Der E,
CTNNB1 activation triggers spermatogonial stem stem cells in mouse testes. Reproduction. 2015;
Yamaji M, Garzia A, Morozov P, Manickavel S,
cell proliferation and germ cell depletion. Dev Biol. 149(3):213–221.
McFarland HL, Roeder RG, Hafner M, Tuschl T.
2017;426(1):17–27. 435. Honecker F, Stoop H, de Krijger RR, Chris Lau YF,
DND1 maintains germline stem cells via re-
419. McIver SC, Stanger SJ, Santarelli DM, Roman SD, Bokemeyer C, Looijenga LH. Pathobiological im-
cruitment of the CCR4–NOT complex to target Nixon B, McLaughlin EA. A unique combination of plications of the expression of markers of testicular
mRNAs. Nature. 2017;543(7646):568–572. male germ cell miRNAs coordinates gonocyte carcinoma in situ by fetal germ cells. J Pathol. 2004;
403. Merlet J, Racine C, Moreau E, Moreno SG, Habert R. differentiation. PLoS One. 2012;7(4):e35553. 203(3):849–857.
Male fetal germ cells are targets for androgens that 420. Takase HM, Nusse R. Paracrine Wnt/b-catenin 436. Rajpert-De Meyts E, Hanstein R, Jørgensen N, Graem
physiologically inhibit their proliferation. Proc Natl signaling mediates proliferation of undifferentiated N, Vogt PH, Skakkebaek NE. Developmental ex-
Acad Sci USA. 2007;104(9):3615–3620. spermatogonia in the adult mouse testis. Proc Natl pression of POU5F1 (OCT-3/4) in normal and
404. Trautmann E, Guerquin MJ, Duquenne C, Lahaye JB, Acad Sci USA. 2016;113(11):E1489–E1497. dysgenetic human gonads. Hum Reprod. 2004;19(6):
Habert R, Livera G. Retinoic acid prevents germ cell 421. Zhou W, Shao H, Zhang D, Dong J, Cheng W, Wang 1338–1344.
mitotic arrest in mouse fetal testes. Cell Cycle. 2008; L, Teng Y, Yu Z. PTEN signaling is required for the 437. Gondos B, Hobel CJ. Ultrastructure of germ cell
7(5):656–664. maintenance of spermatogonial stem cells in development in the human fetal testis. Z Zellforsch
405. DiNapoli L, Batchvarov J, Capel B. FGF9 promotes mouse, by regulating the expressions of PLZF and Mikrosk Anat. 1971;119(1):1–20.
survival of germ cells in the fetal testis. Development. UTF1. Cell Biosci. 2015;5:42. 438. Childs AJ, Cowan G, Kinnell HL, Anderson RA,
2006;133(8):1519–1527. 422. Basciani S, De Luca G, Dolci S, Brama M, Arizzi M, Saunders PT. Retinoic acid signalling and the
406. Olaso R, Habert R. Genetic and cellular analysis of Mariani S, Rosano G, Spera G, Gnessi L. Platelet- control of meiotic entry in the human fetal gonad.
male germ cell development. J Androl. 2000;21(4): derived growth factor receptor b-subtype regulates PLoS One. 2011;6(6):e20249.
497–511. proliferation and migration of gonocytes. Endocri- 439. Gaskell TL, Esnal A, Robinson LL, Anderson RA,
407. Surani MA, Hayashi K, Hajkova P. Genetic and nology. 2008;149(12):6226–6235. Saunders PT. Immunohistochemical profiling of
epigenetic regulators of pluripotency. Cell. 2007; 423. Orth JM, Jester WF Jr, Qiu J. Gonocytes in testes of germ cells within the human fetal testis: identifi-
128(4):747–762. neonatal rats express the c-kit gene. Mol Reprod Dev. cation of three subpopulations. Biol Reprod. 2004;
408. Western PS, van den Bergen JA, Miles DC, Sinclair 1996;45(2):123–131. 71(6):2012–2021.
AH. Male fetal germ cell differentiation involves 424. Orth JM, Qiu J, Jester WF Jr, Pilder S. Expression of 440. Mäkelä JA, Toppari J. Spermatogenic cell syncytium.
complex repression of the regulatory network the c-kit gene is critical for migration of neonatal rat In: Jégou B, Skinner MK, eds. Encyclopedia of Re-
controlling pluripotency. FASEB J. 2010;24(8): gonocytes in vitro. Biol Reprod. 1997;57(3):676–683. production. 2nd ed. New York, NY: Academic Press;
3026–3035. 425. Nurmio M, Toppari J, Zaman F, Andersson AM, 2017:124–133.
409. Kehler J, Tolkunova E, Koschorz B, Pesce M, Gentile Paranko J, Söder O, Jahnukainen K. Inhibition of 441. Ferrara D, Hallmark N, Scott H, Brown R, McKinnell
L, Boiani M, Lomelı́ H, Nagy A, McLaughlin KJ, tyrosine kinases PDGFR and C-Kit by imatinib C, Mahood IK, Sharpe RM. Acute and long-term
Schöler HR, Tomilin A. Oct4 is required for pri- mesylate interferes with postnatal testicular de- effects of in utero exposure of rats to di(n-butyl)
mordial germ cell survival. EMBO Rep. 2004;5(11): velopment in the rat. Int J Androl. 2007;30(4): phthalate on testicular germ cell development
1078–1083. 366–376. and proliferation. Endocrinology. 2006;147(11):
410. Chan AL, La HM, Legrand JMD, Mäkelä JA, 426. Li H, Papadopoulos V, Vidic B, Dym M, Culty M. 5352–5362.
Eichenlaub M, De Seram M, Ramialison M, Hobbs Regulation of rat testis gonocyte proliferation by 442. Hoei-Hansen CE, Almstrup K, Nielsen JE, Brask
RM. Germline stem cell activity is sustained by platelet-derived growth factor and estradiol: Sonne S, Graem N, Skakkebaek NE, Leffers H,

902 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

Rajpert-De Meyts E. Stem cell pluripotency factor 460. Guibert S, Forné T, Weber M. Global profiling of compatible with postnatal development. Dev Cell.
NANOG is expressed in human fetal gonocytes, DNA methylation erasure in mouse primordial 2013;24(3):310–323.
testicular carcinoma in situ and germ cell tumours. germ cells. Genome Res. 2012;22(4):633–641. 476. He YF, Li BZ, Li Z, Liu P, Wang Y, Tang Q, Ding J, Jia Y,
Histopathology. 2005;47(1):48–56. 461. Seki Y, Hayashi K, Itoh K, Mizugaki M, Saitou M, Chen Z, Li L, Sun Y, Li X, Dai Q, Song CX, Zhang K,
443. Forest MG, Cathiard AM, Bertrand JA. Evidence of Matsui Y. Extensive and orderly reprogramming of He C, Xu GL. Tet-mediated formation of 5-car-
testicular activity in early infancy. J Clin Endocrinol genome-wide chromatin modifications associated boxylcytosine and its excision by TDG in mam-
Metab. 1973;37(1):148–151. with specification and early development of germ malian DNA. Science. 2011;333(6047):1303–1307.
444. Forest MG, Sizonenko PC, Cathiard AM, Bertrand J. cells in mice. Dev Biol. 2005;278(2):440–458. 477. Maiti A, Drohat AC. Thymine DNA glycosylase can
Hypophyso-gonadal function in humans during the 462. Hargan-Calvopina J, Taylor S, Cook H, Hu Z, Lee SA, rapidly excise 5-formylcytosine and 5-carbox-
first year of life. 1. Evidence for testicular activity in Yen MR, Chiang YS, Chen PY, Clark AT. Stage- ylcytosine: potential implications for active deme-
early infancy. J Clin Invest. 1974;53(3):819–828. specific demethylation in primordial germ cells thylation of CpG sites. J Biol Chem. 2011;286(41):
445. Hadziselimović F, Thommen L, Girard J, Herzog B. safeguards against precocious differentiation. Dev 35334–35338.
The significance of postnatal gonadotropin surge Cell. 2016;39(1):75–86. 478. Guo F, Yan L, Guo H, Li L, Hu B, Zhao Y, Yong J, Hu Y,
for testicular development in normal and crypt- 463. Kobayashi H, Sakurai T, Miura F, Imai M, Mochiduki Wang X, Wei Y, Wang W, Li R, Yan J, Zhi X, Zhang Y,
orchid testes. J Urol. 1986;136(1 Pt 2):274–276. K, Yanagisawa E, Sakashita A, Wakai T, Suzuki Y, Ito Jin H, Zhang W, Hou Y, Zhu P, Li J, Zhang L, Liu S,
446. Job JC, Toublanc JE, Chaussain JL, Gendrel D, Garnier T, Matsui Y, Kono T. High-resolution DNA meth- Ren Y, Zhu X, Wen L, Gao YQ, Tang F, Qiao J. The
P, Roger M. Endocrine and immunological findings ylome analysis of primordial germ cells identifies transcriptome and DNA methylome landscapes of

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


in cryptorchid infants. Horm Res. 1988;30(4–5): gender-specific reprogramming in mice. Genome human primordial germ cells. Cell. 2015;161(6):
167–172. Res. 2013;23(4):616–627. 1437–1452.
447. Mamsen LS, Lutterodt MC, Andersen EW, Byskov 464. Hajkova P, Ancelin K, Waldmann T, Lacoste N, 479. Kagiwada S, Kurimoto K, Hirota T, Yamaji M, Saitou
AG, Andersen CY. Germ cell numbers in human Lange UC, Cesari F, Lee C, Almouzni G, Schneider R, M. Replication-coupled passive DNA demethyla-
embryonic and fetal gonads during the first two Surani MA. Chromatin dynamics during epigenetic tion for the erasure of genome imprints in mice.
trimesters of pregnancy: analysis of six published reprogramming in the mouse germ line. Nature. EMBO J. 2013;32(3):340–353.
studies. Hum Reprod. 2011;26(8):2140–2145. 2008;452(7189):877–881. 480. Prokopuk L, Stringer JM, Hogg K, Elgass KD, Western
448. Müller J, Skakkebaek NE. Quantification of germ 465. Lane N, Dean W, Erhardt S, Hajkova P, Surani A, PS. PRC2 is required for extensive reorganization of
cells and seminiferous tubules by stereological ex- Walter J, Reik W. Resistance of IAPs to methylation H3K27me3 during epigenetic reprogramming in
amination of testicles from 50 boys who suffered reprogramming may provide a mechanism for mouse fetal germ cells. Epigenetics Chromatin. 2017;
from sudden death. Int J Androl. 1983;6(2):143–156. epigenetic inheritance in the mouse. Genesis. 2003; 10:7.
449. Paniagua R, Nistal M. Morphological and histo- 35(2):88–93. 481. Kim S, Günesdogan U, Zylicz JJ, Hackett JA, Cougot
metric study of human spermatogonia from birth 466. Tam PP, Zhou SX, Tan SS. X-chromosome activity of D, Bao S, Lee C, Dietmann S, Allen GE, Sengupta R,
the mouse primordial germ cells revealed by the Surani MA. PRMT5 protects genomic integrity
to the onset of puberty. J Anat. 1984;139(Pt 3):
expression of an X-linked lacZ transgene. Develop- during global DNA demethylation in primordial
535–552.
ment. 1994;120(10):2925–2932. germ cells and preimplantation embryos. Mol Cell.
450. Chemes HE. Infancy is not a quiescent period of
467. Sugimoto M, Abe K. X chromosome reactivation 2014;56(4):564–579.
testicular development. Int J Androl. 2001;24(1):2–7.
initiates in nascent primordial germ cells in mice. 482. Gomes Fernandes M, He N, Wang F, Van Iperen L,
451. Masliukaite I, Hagen JM, Jahnukainen K, Stukenborg
PLoS Genet. 2007;3(7):e116. Eguizabal C, Matorras R, Roelen BAJ, Chuva De
JB, Repping S, van der Veen F, van Wely M, van Pelt
468. Seki Y, Yamaji M, Yabuta Y, Sano M, Shigeta M, Sousa Lopes SM. Human-specific subcellular com-
AM. Establishing reference values for age-related
Matsui Y, Saga Y, Tachibana M, Shinkai Y, Saitou M. partmentalization of P-element induced wimpy
spermatogonial quantity in prepubertal human
Cellular dynamics associated with the genome-wide testis-like (PIWIL) granules during germ cell de-
testes: a systematic review and meta-analysis. Fertil
epigenetic reprogramming in migrating primordial velopment and spermatogenesis. Hum Reprod.
Steril. 2016;106(7):1652–1657.e2.
germ cells in mice. Development. 2007;134(14): 2018;33(2):258–269.
452. Saitou M, Kagiwada S, Kurimoto K. Epigenetic
2627–2638. 483. Chen Q, Yan W, Duan E. Epigenetic inheritance of
reprogramming in mouse pre-implantation devel-
469. Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee C, acquired traits through sperm RNAs and sperm
opment and primordial germ cells. Development. Down TA, Surani MA. Germline DNA demethy- RNA modifications. Nat Rev Genet. 2016;17(12):
2012;139(1):15–31. lation dynamics and imprint erasure through 5- 733–743.
453. Messerschmidt DM, Knowles BB, Solter D. DNA hydroxymethylcytosine. Science. 2013;339(6118): 484. Davis TL, Yang GJ, McCarrey JR, Bartolomei MS. The
methylation dynamics during epigenetic reprog- 448–452. H19 methylation imprint is erased and re-
ramming in the germline and preimplantation 470. Yamaguchi S, Hong K, Liu R, Inoue A, Shen L, Zhang established differentially on the parental alleles
embryos. Genes Dev. 2014;28(8):812–828. K, Zhang Y. Dynamics of 5-methylcytosine and 5- during male germ cell development. Hum Mol
454. Ferguson-Smith AC. Genomic imprinting: the hydroxymethylcytosine during germ cell reprog- Genet. 2000;9(19):2885–2894.
emergence of an epigenetic paradigm [published ramming. Cell Res. 2013;23(3):329–339. 485. Li JY, Lees-Murdock DJ, Xu GL, Walsh CP. Timing of
correction appears in Nat Rev Genet. 2011;12(9): 471. Vincent JJ, Huang Y, Chen PY, Feng S, Calvopiña JH, establishment of paternal methylation imprints in
663]. Nat Rev Genet. 2011;12(8):565–575. Nee K, Lee SA, Le T, Yoon AJ, Faull K, Fan G, Rao A, the mouse. Genomics. 2004;84(6):952–960.
455. Saitou M, Miyauchi H. Gametogenesis from plu- Jacobsen SE, Pellegrini M, Clark AT. Stage-specific 486. Kobayashi H, Sakurai T, Imai M, Takahashi N,
ripotent stem cells. Cell Stem Cell. 2016;18(6): roles for Tet1 and Tet2 in DNA demethylation in Fukuda A, Yayoi O, Sato S, Nakabayashi K, Hata K,
721–735. primordial germ cells. Cell Stem Cell. 2013;12(4): Sotomaru Y, Suzuki Y, Kono T. Contribution of
456. Tang WW, Kobayashi T, Irie N, Dietmann S, Surani 470–478. intragenic DNA methylation in mouse gametic
MA. Specification and epigenetic programming of 472. Sharif J, Muto M, Takebayashi S, Suetake I, Iwamatsu DNA methylomes to establish oocyte-specific
the human germ line. Nat Rev Genet. 2016;17(10): A, Endo TA, Shinga J, Mizutani-Koseki Y, Toyoda T, heritable marks. PLoS Genet. 2012;8(1):e1002440.
585–600. Okamura K, Tajima S, Mitsuya K, Okano M, Koseki 487. Guo J, Grow EJ, Yi C, Mlcochova H, Maher GJ,
457. Seisenberger S, Andrews S, Krueger F, Arand J, H. The SRA protein Np95 mediates epigenetic in- Lindskog C, Murphy PJ, Wike CL, Carrell DT, Goriely
Walter J, Santos F, Popp C, Thienpont B, Dean W, heritance by recruiting Dnmt1 to methylated DNA. A, Hotaling JM, Cairns BR. Chromatin and single-cell
Reik W. The dynamics of genome-wide DNA Nature. 2007;450(7171):908–912. RNA-seq profiling reveal dynamic signaling and
methylation reprogramming in mouse primordial 473. Yamaguchi S, Shen L, Liu Y, Sendler D, Zhang Y. Role metabolic transitions during human spermatogo-
germ cells. Mol Cell. 2012;48(6):849–862. of Tet1 in erasure of genomic imprinting. Nature. nial stem cell development. Cell Stem Cell. 2017;
458. Tang WW, Dietmann S, Irie N, Leitch HG, Floros VI, 2013;504(7480):460–464. 21(4):533–546.e6.
Bradshaw CR, Hackett JA, Chinnery PF, Surani MA. 474. Yamaguchi S, Hong K, Liu R, Shen L, Inoue A, Diep D, 488. La Salle S, Mertineit C, Taketo T, Moens PB, Bestor
A unique gene regulatory network resets the hu- Zhang K, Zhang Y. Tet1 controls meiosis by reg- TH, Trasler JM. Windows for sex-specific methyla-
man germline epigenome for development. Cell. ulating meiotic gene expression. Nature. 2012; tion marked by DNA methyltransferase expression
2015;161(6):1453–1467. 492(7429):443–447. profiles in mouse germ cells. Dev Biol. 2004;268(2):
459. Smith ZD, Chan MM, Mikkelsen TS, Gu H, Gnirke A, 475. Dawlaty MM, Breiling A, Le T, Raddatz G, Barrasa MI, 403–415.
Regev A, Meissner A. A unique regulatory phase of Cheng AW, Gao Q, Powell BE, Li Z, Xu M, Faull KF, 489. Bourc’his D, Bestor TH. Meiotic catastrophe and
DNA methylation in the early mammalian embryo. Lyko F, Jaenisch R. Combined deficiency of Tet1 retrotransposon reactivation in male germ cells
Nature. 2012;484(7394):339–344. and Tet2 causes epigenetic abnormalities but is lacking Dnmt3L. Nature. 2004;431(7004):96–99.

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 903


REVIEW

490. Kaneda M, Okano M, Hata K, Sado T, Tsujimoto N, in puberty and precedes attainment of the adult 519. Bougnères P, François M, Pantalone L, Rodrigue D,
Li E, Sasaki H. Essential role for de novo DNA complement of undifferentiated spermatogonia. Bouvattier C, Demesteere E, Roger D, Lahlou N.
methyltransferase Dnmt3a in paternal and mater- Reproduction. 2012;143(4):513–522. Effects of an early postnatal treatment of hypo-
nal imprinting. Nature. 2004;429(6994):900–903. 505. Bidlingmaier F, Dörr HG, Eisenmenger W, Kuhnle U, gonadotropic hypogonadism with a continuous
491. Galetzka D, Weis E, Tralau T, Seidmann L, Haaf T. Knorr D. Testosterone and androstenedione con- subcutaneous infusion of recombinant follicle-
Sex-specific windows for high mRNA expression of centrations in human testis and epididymis during stimulating hormone and luteinizing hormone.
DNA methyltransferases 1 and 3A and methyl- the first two years of life. J Clin Endocrinol Metab. J Clin Endocrinol Metab. 2008;93(6):2202–2205.
CpG-binding domain proteins 2 and 4 in human 1983;57(2):311–315. 520. Lambert AS, Bougneres P. Growth and descent of
fetal gonads. Mol Reprod Dev. 2007;74(2):233–241. 506. Berensztein EB, Sciara MI, Rivarola MA, Belgorosky the testes in infants with hypogonadotropic
492. Oakes CC, La Salle S, Smiraglia DJ, Robaire B, Trasler A. Apoptosis and proliferation of human testicular hypogonadism receiving subcutaneous gonado-
JM. Developmental acquisition of genome-wide somatic and germ cells during prepuberty: high rate tropin infusion. Int J Pediatr Endocrinol. 2016;
DNA methylation occurs prior to meiosis in male of testicular growth in newborns mediated by 2016(1):13.
germ cells. Dev Biol. 2007;307(2):368–379. decreased apoptosis. J Clin Endocrinol Metab. 2002; 521. Stoupa A, Samara-Boustani D, Flechtner I, Pinto G,
493. Shirakawa T, Yaman-Deveci R, Tomizawa S, 87(11):5113–5118. Jourdon I, González-Briceño L, Bidet M, Laborde K,
Kamizato Y, Nakajima K, Sone H, Sato Y, Sharif J, 507. Main KM, Toppari J, Suomi AM, Kaleva M, Chevenne D, Millischer AE, Lottmann H, Blanc T,
Yamashita A, Takada-Horisawa Y, Yoshida S, Ura K, Chellakooty M, Schmidt IM, Virtanen HE, Boisen Aigrain Y, Polak M, Beltrand J. Efficacy and safety of
Muto M, Koseki H, Suda T, Ohbo K. An epigenetic KA, Kai CM, Damgaard IN, Skakkebaek NE. Larger continuous subcutaneous infusion of recombinant

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


switch is crucial for spermatogonia to exit the testes and higher inhibin B levels in Finnish than in human gonadotropins for congenital micropenis
undifferentiated state toward a Kit-positive iden- Danish newborn boys. J Clin Endocrinol Metab. 2006; during early infancy. Horm Res Paediatr. 2017;87(2):
tity. Development. 2013;140(17):3565–3576. 91(7):2732–2737. 103–110.
494. Aravin AA, Sachidanandam R, Bourc’his D, Schaefer 508. Kollin C, Granholm T, Nordenskjöld A, Ritzén EM. 522. Boukari K, Meduri G, Brailly-Tabard S, Guibourdenche
C, Pezic D, Toth KF, Bestor T, Hannon GJ. A piRNA Growth of spontaneously descended and surgically J, Ciampi ML, Massin N, Martinerie L, Picard JY, Rey R,
pathway primed by individual transposons is linked treated testes during early childhood. Pediatrics. Lombès M, Young J. Lack of androgen receptor
to de novo DNA methylation in mice. Mol Cell. 2013;131(4):e1174–e1180. expression in Sertoli cells accounts for the absence of
2008;31(6):785–799. 509. Kuijper EA, van Kooten J, Verbeke JI, van Rooijen M, anti-Mullerian hormone repression during early hu-
495. Meikar O, Da Ros M, Korhonen H, Kotaja N. Lambalk CB. Ultrasonographically measured tes- man testis development. J Clin Endocrinol Metab.
Chromatoid body and small RNAs in male germ ticular volumes in 0- to 6-year-old boys. Hum 2009;94(5):1818–1825.
cells. Reproduction. 2011;142(2):195–209. Reprod. 2008;23(4):792–796. 523. Tapanainen JS, Aittomäki K, Min J, Vaskivuo T,
496. Carrieri C, Comazzetto S, Grover A, Morgan M, 510. Tilbrook AJ, Clarke IJ. Negative feedback regulation Huhtaniemi IT. Men homozygous for an inacti-
Buness A, Nerlov C, O’Carroll D. A transit-amplifying of the secretion and actions of gonadotropin- vating mutation of the follicle-stimulating hormone
population underpins the efficient regenerative
releasing hormone in males. Biol Reprod. 2001; (FSH) receptor gene present variable suppression of
capacity of the testis. J Exp Med. 2017;214(6):
64(3):735–742. spermatogenesis and fertility. Nat Genet. 1997;15(2):
1631–1641.
511. Plant TM, Padmanabhan V, Ramaswamy S, 205–206.
497. Orth JM, Gunsalus GL, Lamperti AA. Evidence from
McConnell DS, Winters SJ, Groome N, Midgley AR 524. Chemes HE, Rey RA, Nistal M, Regadera J, Musse
Sertoli cell-depleted rats indicates that spermatid
Jr, McNeilly AS. Circulating concentrations of di- M, González-Peramato P, Serrano A. Physiological
number in adults depends on numbers of Sertoli
meric inhibin A and B in the male rhesus monkey androgen insensitivity of the fetal, neonatal, and
cells produced during perinatal development. En-
(Macaca mulatta). J Clin Endocrinol Metab. 1997; early infantile testis is explained by the ontogeny
docrinology. 1988;122(3):787–794.
82(8):2617–2621. of the androgen receptor expression in Sertoli
498. Johnson L, Zane RS, Petty CS, Neaves WB.
512. Andersson AM, Müller J, Skakkebaek NE. Different cells. J Clin Endocrinol Metab. 2008;93(11):
Quantification of the human Sertoli cell pop-
roles of prepubertal and postpubertal germ cells 4408–4412.
ulation: its distribution, relation to germ cell
and Sertoli cells in the regulation of serum inhibin B 525. McKinnell C, Saunders PT, Fraser HM, Kelnar CJ,
numbers, and age-related decline. Biol Reprod.
levels. J Clin Endocrinol Metab. 1998;83(12): Kivlin C, Morris KD, Sharpe RM. Comparison of
1984;31(4):785–795.
499. Cortes D, Müller J, Skakkebaek NE. Proliferation of 4451–4458. androgen receptor and oestrogen receptor beta
Sertoli cells during development of the human 513. Ramaswamy S, Marshall GR, McNeilly AS, Plant TM. immunoexpression in the testes of the common
testis assessed by stereological methods. Int J Androl. Evidence that in a physiological setting Sertoli cell marmoset (Callithrix jacchus) from birth to adult-
1987;10(4):589–596. number is the major determinant of circulating hood: low androgen receptor immunoexpression in
500. Rey RA, Campo SM, Bedecarrás P, Nagle CA, concentrations of inhibin B in the adult male rhesus Sertoli cells during the neonatal increase in tes-
Chemes HE. Is infancy a quiescent period of tes- monkey (Macaca mulatta). J Androl. 1999;20(3): tosterone concentrations. Reproduction. 2001;
ticular development? Histological, morphometric, 430–434. 122(3):419–429.
and functional study of the seminiferous tubules of 514. Toppari J, Kaipia A, Kaleva M, Laato M, de Kretser 526. Kelsey TW, Miles A, Mitchell RT, Anderson RA,
the cebus monkey from birth to the end of puberty. DM, Krummen LA, Mather JP, Salmi TT. Inhibin Wallace WH. A normative model of serum in-
J Clin Endocrinol Metab. 1993;76(5):1325–1331. gene expression in a large cell calcifying Sertoli cell hibin B in young males. PLoS One. 2016;11(4):
501. Sharpe RM, Walker M, Millar MR, Atanassova N, tumour and serum inhibin and activin levels. APMIS. e0153843.
Morris K, McKinnell C, Saunders PT, Fraser HM. 1998;106(1–6):101–112. 527. Raivio T, Toppari J, Perheentupa A, McNeilly AS,
Effect of neonatal gonadotropin-releasing hormone 515. Rajpert-De Meyts E, Jørgensen N, Graem N, Müller J, Dunkel L. Treatment of prepubertal gonadotrophin-
antagonist administration on Sertoli cell number Cate RL, Skakkebaek NE. Expression of anti- deficient boys with recombinant human follicle-
and testicular development in the marmoset: Müllerian hormone during normal and pathological stimulating hormone. Lancet. 1997;350(9073):263–264.
comparison with the rat. Biol Reprod. 2000;62(6): gonadal development: association with differentiation 528. Young J, Chanson P, Salenave S, Noël M, Brailly S,
1685–1693. of Sertoli and granulosa cells. J Clin Endocrinol Metab. O’Flaherty M, Schaison G, Rey R. Testicular anti-
502. Marshall GR, Plant TM. Puberty occurring either 1999;84(10):3836–3844. Mullerian hormone secretion is stimulated by
spontaneously or induced precociously in rhesus 516. Jeffery A, Streeter AJ, Hosking J, Wilkin TJ, Nelson recombinant human FSH in patients with con-
monkey (Macaca mulatta) is associated with a SM. Anti-Müllerian hormone in children: a ten-year genital hypogonadotropic hypogonadism. J Clin
marked proliferation of Sertoli cells. Biol Reprod. prospective longitudinal study. J Pediatr Endocrinol Endocrinol Metab. 2005;90(2):724–728.
1996;54(6):1192–1199. Metab. 2015;28(9–10):1153–1162. 529. Dwyer AA, Sykiotis GP, Hayes FJ, Boepple PA, Lee H,
503. Simorangkir DR, Marshall GR, Plant TM. Sertoli cell 517. Al-Attar L, Noël K, Dutertre M, Belville C, Forest MG, Loughlin KR, Dym M, Sluss PM, Crowley WF Jr,
proliferation during prepubertal development in Burgoyne PS, Josso N, Rey R. Hormonal and cellular Pitteloud N. Trial of recombinant follicle-stimulating
the rhesus monkey (Macaca mulatta) is maximal regulation of Sertoli cell anti-Müllerian hormone hormone pretreatment for GnRH-induced fertility
during infancy when gonadotropin secretion is production in the postnatal mouse. J Clin Invest. in patients with congenital hypogonadotropic
robust. J Clin Endocrinol Metab. 2003;88(10): 1997;100(6):1335–1343. hypogonadism. J Clin Endocrinol Metab. 2013;
4984–4989. 518. Main KM, Schmidt IM, Toppari J, Skakkebaek NE. 98(11):E1790–E1795.
504. Simorangkir DR, Ramaswamy S, Marshall GR, Early postnatal treatment of hypogonadotropic 530. Ramaswamy S, Plant TM, Marshall GR. Pulsatile
Roslund R, Plant TM. Sertoli cell differentiation in hypogonadism with recombinant human FSH and stimulation with recombinant single chain human
rhesus monkey (Macaca mulatta) is an early event LH. Eur J Endocrinol. 2002;146(1):75–79. luteinizing hormone elicits precocious sertoli cell

904 Mäkelä et al Testis Development Endocrine Reviews, August 2019, 40(4):857–905


REVIEW

proliferation in the juvenile male rhesus monkey thank Lise Akslglaede for sharing the data that were used to forkhead box L2; GADD45g, growth arrest and DNA damage-
(Macaca mulatta). Biol Reprod. 2000;63(1):82–88. produce Figure 7. inducible 45g; GATA, GATA-binding protein; GCT, germ cell
531. Rey R, Lordereau-Richard I, Carel JC, Barbet P, Cate RL, Financial Support: This work was supported by grants tumor; GPT, gonocytes-to-prespermatogonia transition; GR,
Roger M, Chaussain JL, Josso N. Anti-müllerian from the Academy of Finland, the Sigrid Jusélius Foundation, gonadal ridge; GRN, gene regulatory network; GST, gonocytes-
hormone and testosterone serum levels are inversely the Novo Nordisk Foundation, Turku University Hospital, to-spermatogonia transition; GW, gestational week; H2A/
during normal and precocious pubertal develop- and by the Emil Aaltonen Foundation. H4R3me2, dimethylations of arginine 3 on hsitones H2A and
ment. J Clin Endocrinol Metab. 1993;77(5):1220–1226. Correspondence and Reprint Requests: Jorma Toppari, H4; H3K9, histone 3 at lysine 9; H3K27me3, trimethylation of
532. Rey R, Mebarki F, Forest MG, Mowszowicz I, Cate RL, MD, PhD, Institute of Biomedicine, University of Turku, Kii- lysine 27 on histone 3; H3K9me2, dimethylation of lysine
Morel Y, Chaussain JL, Josso N. Anti-müllerian namyllynkatu 10, 20520 Turku, Finland. E-mail: jortop@utu.fi. 9 on histone H3; hESC, human ESC; HH, hedgehog; hiPSC,
Disclosure Summary: The authors have nothing to human-induced pluripotent stem cell; HOX, Homeobox;
hormone in children with androgen insensitivity.
disclose. HPG, hypothalamus–pituitary–gonadal; hPGC, human PGC;
J Clin Endocrinol Metab. 1994;79(4):960–964.
hPGCLC, human PGC-like cell; iMeLC, incipient mesoderm/
533. Sadov S, Koskenniemi JJ, Virtanen HE, Perheentupa
Abbreviations primitive streak-like cell; INSL3, insulin-like peptide 3; ITGB1,
A, Petersen JH, Skakkebaek NE, Main KM, Toppari
5hmC, 5-hydroxymethylcytosine; 5mC, 5-methylcytosine; integrin subunit b1; KDM3A, lysine demethylase 3A; KTS,
J. Testicular growth during puberty in boys with and AGP, adrenogonadal primordium; AMH, anti-Mul̈ lerian lysine-threonine-serine; LH, luteinizing hormone; LHX9, LIM
without a history of congenital cryptorchidism. hormone; AR, androgen receptor; bFGF, basic FGF; BLIMP1, homeobox 9; LIF, leukemia inhibitory factor; MAP3K4, MAPK
J Clin Endocrinol Metab. 2016;101(6):2570–2577. B lymphocyte-induced maturation protein-1; BMP, bone kinase kinase 4; mPGC, mouse PGC; mPGCLC, mPGC-like cell;

Downloaded from https://academic.oup.com/edrv/article/40/4/857/5257800 by guest on 16 February 2024


534. Figueiredo AFA, França LR, Hess RA, Costa GM. morphogenetic protein; CBX2, chromobox homolog 2; CDH1, NR5A1, nuclear receptor subfamily 5 group A member 1;
Sertoli cells are capable of proliferation into cadherin 1; CE, coelomic epithelial/epithelia/epithelium; CIS, OCT4, octamer-binding TF4; PDGF, platelet-derived growth
adulthood in the transition region between the carcinoma in situ; CITED, CREB-binding protein/p300- factor; PDRM14, PR/SET domain 14; PGC, primordial germ cell;
seminiferous tubules and the rete testis in Wistar interacting transactivator, with Glu/Asp-rich carboxyl- PGD2, prostaglandin D2; piRNA, Piwi-interacting RNA; PMC,
rats. Cell Cycle. 2016;15(18):2486–2496. terminal domain; CXCL12, C-X-C motif chemokine ligand peritubular myoid cell; PND, postnatal day; PRDM, PR/SET
535. Perheentupa A, Sadov S, Rönkä R, Virtanen HE, 12; CXCR4, C-X-C motif chemokine receptor 4; CYP26B1, domain; PSC, pluripotent stem cell; PTEN, phosphatase and
Rodprasert W, Vierula M, Jørgensen N, Skakkebæk cytochrome P450 family 26 subfamily B member 1; DAZL, tensin homolog; PTGDS, prostaglandin D2 synthase; RA, reti-
NE, Toppari J. Semen quality improves marginally depleted in azoospermia-like; DDX4, DEAD-box helicase 4; noic acid; ROR2, receptor tyrosine kinase–like orphan receptor
during young adulthood: a longitudinal follow-up DHH, desert hedgehog; DMRT1, doublesex and mab-3–related 2; SCF, stem cell factor; SF1, steroidogenic factor 1; SIX, SIX
study. Hum Reprod. 2016;31(3):502–510. TF1; DND1, DND miRNA-mediated repression inhibitor 1; homeobox; SOX, SRY-box; SRY, sex-determining region Y; SSC,
DNMT, DNA methyltransferase; dpc, days postconception; spermatogonial stem cell; TE, transposable element; TEI,
DPPA, developmental pluripotency-associated protein; DSD, transgenerational epigenetic inheritance; TES, testis-specific
Acknowledgments disorder of sex development; E, embryonic day; ECM, extra- enhancer of Sox9; TET, ten-eleven translocation; TF, tran-
We acknowledge Emmi Rotgers, Anne Jørgensen, and cellular matrix; EMX2, empty spiracles homeobox 2; EOMES, scription factor; TFAP2C, TF AP-2g; TNAP, tissue-nonspecific
Humphrey Yao for sharing their review manuscript titled “At eomesodermin; EpiLC, epiblast-like cell; ESC, embryonic stem alkaline phosphatase; UHRF1, ubiquitin-like with PHD and ring
the Crossroads of Fate—Somatic Cell Lineage Specification in cell; ExE, extraembryonic ectoderm; FGF, fibroblast growth finger domains 1; VEC, vascular endothelial cell; wpc, weeks post
the Fetal Gonad” during finalization of our review. We also factor; FGFR, FGF receptor; FOG2, friend of GATA2; FOXL2, coitum; WT1, Wilms tumor 1..

doi: 10.1210/er.2018-00140 https://academic.oup.com/edrv 905

You might also like