当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 内分泌学杂志 > 2005年 > 第3期 > 正文
编号:11168212
Transcriptional Regulation of Gonadal Development and Differentiation
     Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611

    Address all correspondence and requests for reprints to: J. Larry Jameson, Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611. E-mail: ljameson@northwestern.edu.

    Abstract

    The embryonic gonad is undifferentiated in males and females until a critical stage when the sex chromosomes dictate its development as a testis or ovary. This binary developmental process provides a unique opportunity to delineate the molecular pathways that lead to distinctly different tissues. The testis comprises three main cell types: Sertoli cells, Leydig cells, and germ cells. The Sertoli cells and germ cells reside in seminiferous tubules where spermatogenesis occurs. The Leydig cells populate the interstitial compartment and produce testosterone. The ovary also comprises three main cell types: granulosa cells, theca cells, and oocytes. The oocytes are surrounded by granulosa and theca cells in follicles that grow and differentiate during characteristic reproductive cycles. In this review, we summarize the molecular pathways that regulate the distinct differentiation of these cell types in the developing testis and ovary. In particular, we focus on the transcription factors that initiate these cascades. Although most of the early insights into the sex determination pathway were based on human mutations, targeted mutagenesis in mouse models has revealed key roles for genes not anticipated to regulate gonadal development. Defining these molecular pathways provides the foundation for understanding this critical developmental event and provides new insight into the causes of gonadal dysgenesis.

    LIKE MOST ORGAN systems, mammalian gonadal development involves a complex interplay of multiple cell types, and it occurs during a relatively narrow time window. Thus, it is important to understand the temporal pattern of gene expression as well as the spatial relationships of the developing tissues. For these reasons, most recent studies have used the mouse as a model for examining gonadal development because it is amenable to genetic manipulation as well as detailed histologic and biochemical characterization. During the transition from an undifferentiated gonad to a testis or ovary, the female and male gonad each display characteristic morphological features and patterns of gene expression (1). The coordinated differentiation of the embryonic urogenital ridge, from the bipotential state to sexual dimorphism, provides an opportunity to identify the key factors and commitment steps that underlie gonadal differentiation (2).

    In humans, the functional significance of some of these molecules is evident from mutations that impair gonadal development and reproduction (3, 4, 5). For example, deletions of the Y chromosomal gene, SRY (sex-determining region on the Y chromosome) cause XY male-to-female sex reversal, whereas SRY translocations to the X chromosome lead to XX female-to-male sex reversal (6). The role of Sry as a candidate testis-determining gene was confirmed by demonstrating testis development after transgenic expression of Sry in XX mice (7). Thus, by using animal models, it is possible to explore genetic pathways in greater detail and elucidate disease pathogenesis.

    Many of the gonadal development factors described to date are known to act at the transcriptional level. For the most part, their functions are incompletely understood. Based on homology to other transcription factors, some affect DNA bending (8) or modulate chromatin remodeling (9). Others form interactive complexes that activate transcription (10) or have a role in specifying progenitor cell types (11). Inhibition of gene expression is equally likely to be important as a means to dictate cell fate, but less is known about potential transcriptional repressors. The functional interaction between extracellular ligand molecules and nuclear transcription factors also merits emphasis. For example, extracellular signals can induce transcription factor release and translocation to the nucleus [i.e. ?-catenin/lymphoid-enhancing factor, Smad (a name combining small and mothers against decapentaplegic), Janus kinase/signal transducer and activator of transcription, nuclear factor-B, nuclear factor of activated T cells] (12). Increasing lines of evidence suggest that cross talk among intracellular signaling pathways mediate downstream transcriptional responses. Thus, it is important to identify the extracellular ligands, membrane receptors, and signal transduction pathways associated with gonadal development, as well as the transcription factors.

    To date, numerous candidate genes have been identified based on their pattern of expression in the embryonic gonad. Gonadal phenotypes are increasingly recognized in knockout models originally designed to explore gene function in other tissues. In many cases, embryonic lethality precludes analysis of adult reproductive function. Hence, for gene mutations that have pleiotropic effects, the gonadal phenotype must be studied primarily during embryogenesis, or by cell type-specific removal of a genetic locus. Further identification of novel candidate genes will rely on a combination of molecular and genetic tools, including forward and reverse genetics approaches.

    A model for gonadal development postulates a series of sequential commitment steps as specific cell types achieve their final differentiated state. A goal in the field is to identify the genetic cascade that programs these events. Some of these steps are cell autonomous, meaning that a cell will pursue a particular fate, relatively independent of its environment. More often, key developmental steps occur via cell-to-cell communication, reflecting direct cellular contacts or responses to paracrine signals. Markers of differentiation include morphological changes, as well as alterations in gene or protein expression. A future challenge is to identify specific targets of key genes in the developmental cascade and to characterize the biochemical events associated with differentiation.

    Organogenesis in the Urogenital Ridge

    After gastrulation, organogenesis in the mouse embryo starts approximately 8 d post coitum (dpc). The gonad, adrenal gland, kidney, and reproductive tract can be traced to a derivative of the intermediate mesoderm—the urogenital ridge—that forms at embryonic d 9.5. At 10.5 dpc, the genital ridge thickens bilaterally on either side of the dorsal aorta and constitutes an outgrowth of epithelial cells that proliferate at the center of the coelomic cavity and extend along the anterior-posterior axis from the forelimb bud to the hindlimb bud. The urogenital ridge is tethered to the peritoneal cavity by mesentery and comprises two juxtaposed tissues: the gonad rudiment is located medially and the mesonephros, which contains dual Wolffian and Müllerian ducts, is more dorsal (Fig. 1, A and B). The metanephric kidney is an outgrowth of the caudal mesonephros and is visible by 12.5 dpc (Fig. 1B). The adreno-gonad primordium arises between the gonad and mesonephros on the dorsal side and buds anteriorly at 12.5 dpc to form the adrenal gland. In both sexes, the adrenal gland and kidney ascend toward the abdominal region. In contrast, the final position of the gonad depends on its differentiation into an ovary or testis, and on the interdependent maturation of the sex-specific Müllerian or Wolffian ducts. In the male, owing the action of testosterone, the proximal Wolffian duct is virilized to form the epididymis, vas deferens, and seminal vesicle. Regression of the distal Müllerian duct is mediated by anti-Müllerian hormone (AMH, or Müllerian inhibitory substance, MIS) (13). Another peptide hormone, insulin-like 3 (Insl-3) mediates testicular descent (13, 14). Persistent Müllerian duct syndrome (PMDS) in males is caused by loss of MIS, MIS type I, or type II receptor function (15, 16, 17). The absence of testosterone and MIS in females allows regression of the Wolffian duct and development of the Müllerian duct into oviduct, uterus, and upper vagina.

    FIG. 1. At 11.5 dpc (A), the bipotential urogenital ridge (large arrow) flanks the dorsal aorta (small arrow) along the anterior-posterior axis in between the forelimb bud (FL) and hindlimb bud (HL). The gonad is closer to the midline, and the mesonephros develops dorso-lateral to the gonad. At 11.5 dpc, there are approximately 18 tail somites (ts) from the tip of the tail to the posterior end of the hindlimb bud. B, At 12.5 dpc, the male gonad shows distinct morphologic features including a prominent coelomic blood vessel (arrow). G, Gonad; M, mesonephros; K, kidney. C, By 13.5 dpc, the male gonad (left) is approximately twice the size of the female (right) due to increased cellular proliferation. D, Sexual dimorphism between the male (left) and female (right) gonad is further evident by 14.5 dpc (scale bars, 500 μm).

    Genetic Program of the Bipotential Gonad

    The gonad primordium is morphologically indistinguishable from 10.0 through 11.5 dpc in males and females. The testis cords become visible by 12.0 dpc and reflect the coalescence of Sertoli and germ cells that are surrounded by a layer of thin peritubular myoid cells. In males, the gonad begins to enlarge visibly at about 13.5 dpc, as a consequence of proliferation of germ cells and precursor somatic cells. The ovary, on the other hand, is seemingly more quiescent during this time, but female-specific markers provide evidence that ovarian differentiation is underway (18, 19).

    Before expression of the male determining gene, Sry at 10.5 dpc, a number of factors act in concert to specify the urogenital ridge in both sexes (Fig. 2). Moreover, because the urogenital ridge is the primordium for the gonad, adrenal, kidney, and reproductive tract, multiple organs are often affected by mutations of these genes.

    FIG. 2. Summary of genetic pathways involved in gonadal development. The bipotential gonad expresses genes such as Wt1, Sf1, Emx2, M33, Lhx9, Pod1, and Dmrt1. Subsequently, expression of the insulin receptor family Ir, Irr, and Igf1r; GATA4/Fog2; Wt1; and Sf1 are thought to promote the expression of Sry in the male. Sox9 and Sox8 are downstream of Sry. Sf1 regulates Amh activation in Sertoli cells and steroidogenesis in Leydig cells. In the male, Dax1 may repress Sf1 in a dose-dependent manner, modulating the transactivation of Amh and genes necessary for steroid hormone biosynthesis. Dax1 also supports the normal formation of testis cords. Additional factors such as Arx, Dhh, Pdgfr-, Fgf9, and FgfR2 are involved in Leydig and Sertoli cell differentiation. A description of the cellular events downstream of Sry and thus specific to male development is outlined in the center box. PGC, Primordial germ cell; PTM, peritubular myoid cell. Female differentiation requires the activity of Wnt4a and repression of Sox9. Other key factors include Fst, Bmp2, and Dax1.

    Wt1 (Wilms’ tumor-1) encodes a zinc finger transcription factor originally identified as a tumor suppressor gene. Loss-of-function mutations cause three distinct pediatric syndromes that display urogenital defects affecting male development: WAGR (Wilms’ tumor, aniridia, genitourinary abnormality, mental retardation), Denys-Drash syndrome, and Frasier syndrome. The Wt1 homozygous knockout animal does not form kidney, adrenal, or gonad (20, 21). In the gonad, the expression of Wt1 at embryonic d 9.5 suggests a role in specifying the coelomic epithelial cells in the developing urogenital ridge and ensuring their survival (22). Two major isoforms have relevance to the developing urogenital organs: one alternatively spliced variant contains amino acids KTS (lysine-threonine-serine) (+KTS isoform), and the other lacks KTS (–KTS isoform). The additional amino acids lend flexibility to the protein between the third and fourth zinc finger and thereby diminish its DNA binding property. Both isoforms appear to have nonredundant functions in kidney and gonad development but not in the adrenal (23). In animals with genetically engineered ablation of the –KTS isoform, apoptosis was observed in streak gonads. Sry expression is reduced in the gonads of animals lacking the +KTS splice variant. Loss of either splice variant impairs testis descent.

    Wt1 functions upstream of two orphan nuclear receptors Sf1 (steroidogenic factor 1) and Dax1 (dosage-sensitive sex reversal, adrenal hypoplasia congenita, X chromosome) (11). Wt1 and Sf1 synergize to enhance transcription of Amh, whereas in vitro experiments demonstrate this interaction is repressed by Dax1 (10). Sf1 also regulates the expression of multiple genes involved in male differentiation, steroidogenesis, and reproduction through Sf1-responsive DNA elements in target gene promoters [Ref.24 ; and see review in this issue by Hammer et al. (25)]. Sf1 stimulates Dax1 transcription, and the two receptors are coexpressed in the adrenal, gonad, pituitary gonadotropes, and ventromedial hypothalamus (26). Dax1 interacts directly with Sf1 and functions as a transcriptional repressor of Sf1-regulated genes (27). In Sf1 null mutants, neither XX nor XY animals form the adrenal or gonad. In the absence of Sf1, cells in both tissue rudiments undergo apoptosis, indicating that Sf1 is necessary for survival of early progenitors of the adrenal and gonad (28). At 11.5 dpc, Sf1 colocalizes with proliferating cells, suggesting that it directly or indirectly stimulates cell proliferation (29). Dax1-null males possess gonadal defects in testis cord morphogenesis, peritubular myoid cell proliferation, and spermatogenesis (30, 31). Hence, Sf1 and Dax1 are independently important for normal male gonadal differentiation.

    Genes that primarily affect male gonadal differentiation or, have a male-specific expression pattern before distinct morphological changes, continue to expand the network of molecular pathways involved in testis development. Autosomal recessive male-to-female sex reversal (XY male bearing female gonads and feminized external genitalia) has been described in several mouse knockout models. M33 (polycomb homolog) is involved in chromatin remodeling, yet its precise molecular role in gonadal differentiation is unknown (9). The LIM-domain containing transcription factor Lhx9 (Lim homeobox) is required for full Sf1 expression (11, 32). In contrast to the Sf1 null animal, lack of testis development was not attributed to apoptosis in either Lhx9 or M33 mutant mice. Pod1 (podocyte) is a basic helix-loop-helix transcription factor with a primary role in glomerular visceral epithelial cell (podocyte) differentiation and branching morphogenesis in the kidney and lung (33). In Pod1 knockout mice, the gonads of both sexes are hypoplastic (34). Expression of Pod1 in the male gonad was found mainly in the interstitium: peritubular myoid cells, pericytes associated with endothelium, and fetal Leydig cells. Defective coelomic vessel formation and attenuation of migrating endothelial cells from the mesonephros compromised vascularization in Pod1 knockout male gonads. Although Sertoli development was not affected, there appeared to be expansion of the Sf1-positive Leydig cell population. Dmrt1 (doublesex- and mab-related transcription factor) is homologous to genes in Drosophila and Caenorhabditis elegans, and null mutation causes dysmorphic testes in mice (35). Maestro (Mro), protease nexin-1 (Pn-1), and vanin-1 (Vn-1) were identified based on a sexually dimorphic expression pattern specific to the XY bipotential gonad (36, 37, 38). Later in development, maestro remains expressed in Sertoli and XY germ cells, whereas protease nexin-1 and vanin-1 are Sertoli restricted. Whether any of these newly described factors specify precursor populations, analogous to the role of Sry-expressing Sertoli precursor cells, remains to be tested. The extent to which function can be attributed to a gene product relies on the phenotype of null mutation models in which are affected on all tissues where the gene of interest is expressed. Only recently have tissue-specific mutagenesis studies, for example with Sf1 and Sox9 [Sry related, HMG (high-mobility group) box], begun to elucidate spatial requirements attributed to developmentally regulated genes (39, 40, 41).

    Supporting Cells: The Embryonic Testis Cord

    The Sertoli cell is positioned at the base of the testis cord creating the perimeter of the tubule. Closely apposed is the elongated cell layer of peritubular myoid cells circumferentially surrounding the nascent cord. Testis cord morphogenesis is apparent in the male as early as embryonic d 12.0. The testis cords pattern the developing gonad and are the embryologic foundation for the adult seminiferous tubule. Sertoli precursor cells were identified at the coelomic surface (ventro-medial aspect) using dye tracking experiments that labeled dividing cells (42). Sertoli cells and granulosa cells may originate from the same precursor (43). In XX-XY chimeric studies, Sertoli cells were the only cell type that required Sry expression in a cell autonomous manner, whereas XX cells were preferentially excluded from the Sertoli population (44).

    Sertoli cells, the cell lineage in which Sry is solely expressed in the male gonad, are thought to direct the differentiation of other cell types (45). For example, fetal Leydig cell differentiation depends on paracrine signaling through the extracellular protein Desert Hedgehog, which is secreted by Sertoli cells (46). Endothelial cells originating from the mesonephros are induced by a chemoattractant released by the male gonad (47). Two factors that can induce mesonephric cell migration are fibroblast growth factor (FGF) 9 and MIS, both of which are produced by Sertoli cells (48, 49). Sertoli cells also secrete a postulated factor(s) that induces mitotic arrest of germ cells (50). Unlike the female gonad, germ cells in the developing testis undergo mitotic arrest, being held as diploid progenitors until later in spermatogenesis. There is recent evidence that XY germ cells synthesize paracrine factors, including prostaglandin D2, that induce Sertoli differentiation (51).

    Sry expression occurs in a narrow temporal window from 10.5–12.0 dpc. The spatial pattern of Sry expression has been clarified by in situ hybridization analysis timed to tail somite stages. The central region of the gonad exhibits Sry expression first, followed by a rostral to caudal progression along the entire length of the gonad (52). Although Sry belongs to the HMG box containing family of nuclear transcription factors, evidence for downstream target genes remains elusive. A closely related gene, Sox9, is the other definitive male-determining gene identified thus far. Immunohistochemical labeling of genetically marked Sry-positive cells demonstrate that Sry and Sox9 expression overlap in cells of the Sertoli lineage (53). As the Sry signal diminishes at 12.5 dpc, Sox9 expression increases in the male and reaches a peak at 14.5 dpc. Sox9, which is weakly expressed in the undifferentiated gonad, is down-regulated in the female. Transgenic expression of Sox9 on an XX genetic background is sufficient to cause female-to-male sex reversal (54). Heterozygous human SOX9 mutations cause campomelic dysplasia, a severe skeletal disorder with defective cartilage development (55, 56). Many of these male patients also have gonadal dysgenesis. Heterozygous mice haploinsufficient for Sox9 die perinatally due to skeletal malformations (57). An ingenious strategy was designed to transmit the knockout allele through the germline of otherwise wild-type male and female parents by the use of spermatocyte- and oocyte-specific promoters, respectively, enabling the zygote to obtain both knockout alleles (41). These experiments confirm that Sox9 is necessary for male gonadal development. Notably, Sox9 null embryos have elevated Sry expression, suggesting a negative feedback regulatory loop that down-regulates Sry in the mouse. Experimental evidence using mouse strains with a variety of Sry alleles supports the assertion that the precise timing, duration, threshold transcript levels, and protein isoform half-life, influence downstream events dependent on Sry expression (58, 59). Sox8, another member of the Sox HMG box containing transcription factor gene family, is also active in male development (60). It is possible that Sry, Sox9, and Sox8 have partially redundant functions.

    GATA4 and Fog2 (friend of GATA) are important for cardiac morphogenesis but also influence gonadal development. A GATA4 knock-in mutation, which prevents nuclear association of GATA4 and Fog2, eliminates expression of the male differentiation markers Sox9 and Amh. Fog2 null mice have decreased Sry expression, loss of Sox9, Amh, and Desert hedgehog (Dhh), but persistence of the female marker Wnt4 (61). The GATA4 knock-in and Fog2 null mice retain Sf1 and Wt1 expression, suggesting a hierarchical network. Similarly, Ir, Irr, Igf1r (insulin receptor, insulin-related receptor, Igf1 receptor) triple mutants have low Sry and Sox9 expression and exhibit male-to-female sex reversal, thus implicating the insulin signaling pathway in male differentiation (62).

    Anti-Müllerian hormone (AMH) or MIS is a key peptide hormone produced by Sertoli cells. It mediates the regression of the Müllerian duct, which would otherwise form female reproductive tract structures. In genetic males, PMDS is due to loss of MIS or cognate receptors present in mesenchymal cells of the Müllerian duct. PMDS is an example of male pseudohermaphroditism whereby males with testicular tissue appear feminized externally due to failure of testis descent caused by the presence of uterus and oviduct. Müllerian regression involves activation of a ?-catenin-dependent pathway in a rostral-to-caudal manner (63). A number of factors are known to regulate Amh transactivation. These include Sf1, WT1, GATA4, Sox9, and Sox8 (10, 64, 65, 66). MIS may have other functions that include mesonephric cell migration and vascularization in the male gonad, and germ cell loss in the ovary (49, 67).

    Proliferation of pre-Sertoli cells is an important event in male development (29). By 13.5 dpc, the male gonad is twice the size of the female (see Fig. 1, C and D). One factor linked to proliferation is Fgf9. Fgf9 null mice show varying degrees of male-to-female sex reversal (48). A number of Fgf receptors have been identified and FgfR2 is thought to direct Sertoli cell differentiation perhaps as a result of nuclear translocation, which occurs simultaneously with translocation of Sry and Sox9 (68). A secreted protein, Fgf9, is a candidate male gonad-specific chemoattractant signal that induces migration of cells from the mesonephros into the gonad. Immigrant cells have been identified mainly as endothelial and peritubular myoid cells (47). Migration of these cells is necessary for testis cord morphogenesis (69) and induction of Sox9 expression (70). This migration of mesonephric cells does not occur in females presumably because a chemoattractant is absent. Furthermore, once XX germ cells enter meiosis at 13.5 dpc, migration from the mesonephros is inhibited (71).

    Hormone Biosynthesis: The Interstitial Space

    Once sex determination has occurred, the ensuing phenotypic differentiation depends heavily on the production of steroid hormones. In the male, testosterone stabilizes the Wolffian duct and is converted by 5-reductase to the potent derivative, dihydrotestosterone, which induces virilization of the male external genitalia. In the female, aromatase mediates estradiol synthesis. In some species, aromatase expression is critical for ovarian development; its role in ovary development in mammals is less clear, although aromatase deficiency is associated with transdifferentiation of granulosa cells into Sertoli-like cells (72).

    Sf1 is restricted to Leydig cells after 13.5 dpc and mediates expression of several genes encoding enzymes required for testosterone biosynthesis including StAR (steroidogenic acute regulatory protein), Cyp11a1, Cyp17 (cytochrome P450 hydroxylases), and 3?HSD (hydroxysteroid dehydrogenase). The factors required for Leydig cell determination and lineage development are incompletely understood. Moreover, their origin, whether from immigrant mesonephric cells or progenitors within the gonad, remains equivocal. Leydig cell fate appears to rely on paracrine signals. Dhh, a Sertoli-secreted factor, induces expression of Cyp11a1 (side-chain cleavage) in the Leydig cell (46). The Dhh signal is received by the receptor patched (Ptc1), which is expressed on Leydig cells. Dhh is thought to play a crucial role in Leydig cell differentiation and is an example of a paracrine signaling pathway identified between two developing cell types. Genetic analysis has placed Pdgfr- (platelet-derived growth factor receptor-) upstream of Dhh (73). Like Fgf9, Pdgfs induce mesonephric migration, and Pdgfr- is necessary in the gonad for full Cyp11a1 expression. The X-linked gene Arx (Aristaless-related homeobox) also influences Leydig cell development. However, it is expressed mainly in peritubular myoid cells, endothelial cells, and in the epithelium of the tunica albuginea (74).

    Female Embryonic Gonadal Development

    Relatively few genes have been shown to exhibit a female-specific pattern of gene expression early in gonadal development. To date, no gene has been shown to be a female-determining gene, as defined by conferring complete female-to-male sex reversal when mutated on an XX background, or by conferring male-to-female sex reversal after overexpression on an XY background. The Dax1 gene was initially suggested as a pro-ovarian, or anti-testis, candidate gene because its duplication on an XY background is associated with impaired testis development (75, 76). However, Dax1 loss of function on the XX background does not prevent ovary development (31). Subsequent studies have shown an unexpected role for Dax1 in testis development (77), indicating that its actions are highly dependent on the timing and level of expression.

    Although male somatic tissue can survive in the absence of germ cells, the female gonad depends on migratory germ cells to populate the undifferentiated primordium (78). In the female gonad, germ cells begin entering meiosis at 13.5 dpc and initiate a dynamic paracrine relationship with supporting cells destined to become cumulus and granulosa cells (79).

    A growing list of genes exhibit an ovary-specific expression pattern. For example, follistatin (Fst) is highly expressed in the developing ovary relative to the testis (19). Follistatin binds members of the activin/TGF?/bone morphogenetic protein (BMP) family. Thus, it may regulate the activity of one or more of these factors. Stra8 (stimulated by retinoic acid) is also selectively expressed in the developing ovary and is thought to play a role in regulation of meiosis (80). Targeted mutagenesis and transgenic overexpression experiments will clarify the functions of these factors.

    There has been postulated a "Z factor" that could act to suppress pro-testis events carried out by autosomal or X-linked genes in both XY and XX backgrounds (81). The Z factor would act downstream of Sry in genetic males. Therefore, the Z factor would be repressed by Sry in the male, and independent of Sry on an XX genetic background. Loss of a Z factor would be sex-reversing on XX (female-to-male), but gain-of-function on XY may or may not exhibit sex reversal (male-to-female) depending on whether the Z factor can override the downstream events of Sry. One candidate for the Z factor is Wnt4 (Wingless-related integration site). Female Wnt4 null mice develop virilizing gonads and Wolffian derivatives (82). This gonadal phenotype reflects the action of Wnt4 to induce the migration of steroidogenic precursors of the adreno-gonad primordium into the anterior region of the gonad (83). Hence, misdirected adrenal steroidogenic cells become localized in the XX Wnt4 knockout gonad. Secondly, Wnt4 is down-regulated in males, whereas its expression remains strong in females where it prevents coelomic vascularization (a male feature, see Fig. 1B) through a molecular pathway that involves Fst and Bmp2 (19). By inhibiting vascularization, Wnt4 may prevent the proliferative growth expansion characteristic of male development. Thus, the homozygous knockout female gonad appears somewhat more male-like in shape and size. Externally, however, homozygous knockouts exhibit female genitalia. In humans, a loss-of-function mutation in WNT4 caused Mayer-Rokitansky-Kuster-Hauser syndrome, which is characterized by defective development of Müllerian derivatives (84). Duplication of a chromosomal region containing WNT4 was associated with a case of human XY sex reversal (85). However, overexpression of Wnt4 on an XY background does not lead to XY sex reversal in the mouse (83, 86). Thus, the major roles of Wnt4 in the female appear to include suppression of androgen-producing interstitial cells, inhibition of testis-like vascularization, and support of Müllerian derivatives. Wnt4 is also essential for kidney (87), mammary gland (88), and pituitary development (89), as well as normal functioning of the adrenal cortex (90).

    A dominant insertional mutation led to identification of a female-to-male sex reversal phenotype in the Odsex (Ocular degeneration with sex reversal) mouse (91). The Odsex mouse was initially thought to lack a repressor for Sox9, allowing unabated Sox9 expression ectopically in genetic females. Therefore, one of the functions of a Z factor could be to suppress Sox9 action in the female gonad. However, it is also possible that regulatory elements contained in the inserted transgene promote activation of Sox9.

    Nucleo-cytoplasmic shuttling of transcription factors may also provide an important regulatory step in sex determination. Organ culture experiments using the nuclear export blocker, leptomycin B, resulted in male-characteristic Sox9 and MIS expression in XX gonad organ cultures (92). Thus, growth factors may be able to modulate transcriptional control by altering the location as well as the activity of various transcription factors.

    Central Questions and Future Directions

    The molecular pathways directing cellular differentiation can be traced back to cells of pluripotential origin. Thus, development must occur through a series of commitment steps driven by cell-to-cell contacts and paracrine interactions. In some cases, committed cells initiate a cell-autonomous program, as perhaps illustrated by Sry and Sox9 actions in Sertoli cells. In other cases, differentiation is driven by secreted factors as illustrated by Dhh, Wnt4, and Fgf9. These cell autonomous and paracrine pathways are not mutually exclusive, and it is likely that most commitment steps integrate a series of internal and external signals. The sex determination pathway provides a unique paradigm for considering these events because the commitment steps lead to distinct cell types within a narrow time frame.

    Despite impressive progress in this field, a number of challenging questions remain. What factors initiate Sry expression and what are its cellular targets? Are Sry and Sox9 expression linked directly or do they reflect parallel, partially redundant pathways? Given that Sox9 is sufficient to induce Sertoli cell development, what are its targets and how does it induce a network of genes they convey morphological changes characteristic of the testis? What are the genetic and molecular events that dictate ovary development? Although it is known that germ cells interact actively with somatic cells, what are the molecular and cellular events that control germ cell replication and entry into meiosis? How do meiotic germ cells foster ovary development? Implicit in these questions is the critical feature of timing during gonadal development. Once the Sertoli program is initiated, the developing testis induces mitotic arrest of germ cells. On the other hand, if Sertoli development is delayed, as occurs with various Sry alleles and mouse genetic backgrounds, germ cells begin to enter meiosis, and this step may further impair testis development, leading to mixed testis/ovary tissue or gonadal dysgenesis. Thus, future studies need to carefully assess pathways that control the timing and level of expression of regulatory factors.

    Further understanding of these molecular and cellular functions in the gonad will provide examples of paradigms used to control cellular differentiation in other tissues. This knowledge will also be useful for the characterization of intersex cases and perhaps additional causes of idiopathic infertility. Parallel investigation of human mutations and animal models can accelerate progress in this field by identifying candidate genes and clarifying their roles in development.

    References

    Koopman P, Bullejos M, Loffler K, Bowles J 2002 Expression-based strategies for discovery of genes involved in testis and ovary development. Novartis Found Symp 244:240–257

    Brennan J, Capel B 2004 One tissue, two fates: molecular genetic events that underlie testis versus ovary development. Nat Rev Genet 5:509–521

    Achermann JC, Ozisik G, Meeks JJ, Jameson JL 2002 Genetic causes of human reproductive disease. J Clin Endocrinol Metab 87:2447–2454

    MacLaughlin DT, Donahoe PK 2004 Sex determination and differentiation. N Engl J Med 350:367–378

    Koopman P 2001 The genetics and biology of vertebrate sex determination. Cell 105:843–847

    Harley VR, Clarkson MJ, Argentaro A 2003 The molecular action and regulation of the testis-determining factors, SRY (sex-determining region on the Y chromosome) and SOX9 [SRY-related high-mobility group (HMG) box 9]. Endocr Rev 24:466–487

    Koopman P, Gubbay J, Vivian N, Goodfellow P, Lovell-Badge R 1991 Male development of chromosomally female mice transgenic for Sry. Nature 351:117–121

    Phillips NB, Nikolskaya T, Jancso-Radek A, Ittah V, Jiang F, Singh R, Haas E, Weiss MA 2004 Sry-directed sex reversal in transgenic mice is robust with respect to enhanced DNA bending: comparison of human and murine HMG boxes. Biochemistry 43:7066–7081

    Katoh-Fukui Y, Tsuchiya R, Shiroishi T, Nakahara Y, Hashimoto N, Noguchi K, Higashinakagawa T 1998 Male-to-female sex reversal in M33 mutant mice. Nature 393:688–692

    Nachtigal MW, Hirokawa Y, Enyeart-VanHouten DL, Flanagan JN, Hammer GD, Ingraham HA 1998 Wilms’ tumor 1 and Dax-1 modulate the orphan nuclear receptor SF-1 in sex-specific gene expression. Cell 93:445–454

    Wilhelm D, Englert C 2002 The Wilms tumor suppressor WT1 regulates early gonad development by activation of Sf1. Genes Dev 16:1839–1851

    Xu L, Massague J 2004 Nucleocytoplasmic shuttling of signal transducers. Nat Rev Mol Cell Biol 5:209–219

    di Clemente N, Josso N, Gouedard L, Belville C 2003 Components of the anti-Mullerian hormone signaling pathway in gonads. Mol Cell Endocrinol 211:9–14

    Nef S, Parada LF 1999 Cryptorchidism in mice mutant for Insl3. Nat Genet 22:295–299

    Behringer RR, Finegold MJ, Cate RL 1994 Mullerian-inhibiting substance function during mammalian sexual development. Cell 79:415–425

    Mishina Y, Rey R, Finegold MJ, Matzuk MM, Josso N, Cate RL, Behringer RR 1996 Genetic analysis of the Mullerian-inhibiting substance signal transduction pathway in mammalian sexual differentiation. Genes Dev 10:2577–2587

    Jamin SP, Arango NA, Mishina Y, Hanks MC, Behringer RR 2002 Requirement of Bmpr1a for Mullerian duct regression during male sexual development. Nat Genet 32:408–410

    Bullejos M, Bowles J, Koopman P 2002 Extensive vascularization of developing mouse ovaries revealed by caveolin-1 expression. Dev Dyn 225:95–99

    Yao HH, Matzuk MM, Jorgez CJ, Menke DB, Page DC, Swain A, Capel B 2004 Follistatin operates downstream of Wnt4 in mammalian ovary organogenesis. Dev Dyn 230:210–215

    Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, Housman D, Jaenisch R 1993 WT-1 is required for early kidney development. Cell 74:679–691

    Moore AW, McInnes L, Kreidberg J, Hastie ND, Schedl A 1999 YAC complementation shows a requirement for Wt1 in the development of epicardium, adrenal gland and throughout nephrogenesis. Development 126:1845–1857

    Armstrong JF, Pritchard-Jones K, Bickmore WA, Hastie ND, Bard JB 1993 The expression of the Wilms’ tumour gene, WT1, in the developing mammalian embryo. Mech Dev 40:85–97

    Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, Ziegler U, Gubler MC, Schedl A 2001 Two splice variants of the Wilms’ tumor 1 gene have distinct functions during sex determination and nephron formation. Cell 106:319–329

    Achermann JC, Meeks JJ, Jameson JL 2001 Phenotypic spectrum of mutations in DAX-1 and SF-1. Mol Cell Endocrinol 185:17–25

    Hammer GD, Parker KL, Schimmer BP 2005 Minireview: transcriptional regulation of adrenocortical development. Endocrinology 146:1018–1024

    Ikeda Y, Swain A, Weber TJ, Hentges KE, Zanaria E, Lalli E, Tamai KT, Sassone-Corsi P, Lovell-Badge R, Camerino G, Parker KL 1996 Steroidogenic factor 1 and Dax-1 colocalize in multiple cell lineages: potential links in endocrine development. Mol Endocrinol 10:1261–1272

    Ito M, Yu R, Jameson JL 1997 DAX-1 inhibits SF-1-mediated transactivation via a carboxy-terminal domain that is deleted in adrenal hypoplasia congenita. Mol Cell Biol 17:1476–1483

    Luo X, Ikeda Y, Parker KL 1994 A cell-specific nuclear receptor is essential for adrenal and gonadal development and sexual differentiation. Cell 77:481–490

    Schmahl J, Eicher EM, Washburn LL, Capel B 2000 Sry induces cell proliferation in the mouse gonad. Development 127:65–73

    Meeks JJ, Crawford SE, Russell TA, Morohashi KI, Weiss J, Jameson JL 2003 Dax1 regulates testis cord organization during gonadal differentiation. Development 130:1029–1036

    Yu RN, Ito M, Saunders TL, Camper SA, Jameson JL 1998 Role of Ahch in gonadal development and gametogenesis. Nat Genet 20:353–357

    Birk OS, Casiano DE, Wassif CA, Cogliati T, Zhao L, Zhao Y, Grinberg A, Huang S, Kreidberg JA, Parker KL, Porter FD, Westphal H 2000 The LIM homeobox gene Lhx9 is essential for mouse gonad formation. Nature 403:909–913

    Quaggin SE, Schwartz L, Cui S, Igarashi P, Deimling J, Post M, Rossant J 1999 The basic-helix-loop-helix protein pod1 is critically important for kidney and lung organogenesis. Development 126:5771–5783

    Cui S, Ross A, Stallings N, Parker KL, Capel B, Quaggin SE 2004 Disrupted gonadogenesis and male-to-female sex reversal in Pod1 knockout mice. Development 131:4095–4105

    Raymond CS, Murphy MW, O’Sullivan MG, Bardwell VJ, Zarkower D 2000 Dmrt1, a gene related to worm and fly sexual regulators, is required for mammalian testis differentiation. Genes Dev 14:2587–2595

    Bowles J, Bullejos M, Koopman P 2000 A subtractive gene expression screen suggests a role for vanin-1 in testis development in mice. Genesis 27:124–135

    Smith L, Van Hateren N, Willan J, Romero R, Blanco G, Siggers P, Walsh J, Banerjee R, Denny P, Ponting C, Greenfield A 2003 Candidate testis-determining gene, Maestro (Mro), encodes a novel HEAT repeat protein. Dev Dyn 227:600–607

    Grimmond S, Van Hateren N, Siggers P, Arkell R, Larder R, Soares MB, de Fatima Bonaldo M, Smith L, Tymowska-Lalanne Z, Wells C, Greenfield A 2000 Sexually dimorphic expression of protease nexin-1 and vanin-1 in the developing mouse gonad prior to overt differentiation suggests a role in mammalian sexual development. Hum Mol Genet 9:1553–1560

    Jeyasuria P, Ikeda Y, Jamin SP, Zhao L, De Rooij DG, Themmen AP, Behringer RR, Parker KL 2004 Cell-specific knockout of steroidogenic factor 1 reveals its essential roles in gonadal function. Mol Endocrinol 18:1610–1619

    Zhao L, Bakke M, Parker KL 2001 Pituitary-specific knockout of steroidogenic factor 1. Mol Cell Endocrinol 185:27–32

    Chaboissier MC, Kobayashi A, Vidal VI, Lutzkendorf S, van de Kant HJ, Wegner M, de Rooij DG, Behringer RR, Schedl A 2004 Functional analysis of Sox8 and Sox9 during sex determination in the mouse. Development 131:1891–1901

    Karl J, Capel B 1998 Sertoli cells of the mouse testis originate from the coelomic epithelium. Dev Biol 203:323–333

    Albrecht KH, Eicher EM 2001 Evidence that Sry is expressed in pre-Sertoli cells and Sertoli and granulosa cells have a common precursor. Dev Biol 240:92–107

    Palmer SJ, Burgoyne PS 1991 In situ analysis of fetal, prepuberal and adult XX-XY chimaeric mouse testes: Sertoli cells are predominantly, but not exclusively, XY. Development 112:265–268

    Swain A, Lovell-Badge R 1999 Mammalian sex determination: a molecular drama. Genes Dev 13:755–767

    Yao HH, Whoriskey W, Capel B 2002 Desert hedgehog/patched 1 signaling specifies fetal Leydig cell fate in testis organogenesis. Genes Dev 16:1433–1440

    Martineau J, Nordqvist K, Tilmann C, Lovell-Badge R, Capel B 1997 Male-specific cell migration into the developing gonad. Curr Biol 7:958–968

    Colvin JS, Green RP, Schmahl J, Capel B, Ornitz DM 2001 Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell 104:875–889

    Ross AJ, Tilman C, Yao H, MacLaughlin D, Capel B 2003 AMH induces mesonephric cell migration in XX gonads. Mol Cell Endocrinol 211:1–7

    McLaren A, Southee D 1997 Entry of mouse embryonic germ cells into meiosis. Dev Biol 187:107–113

    Adams IR, McLaren A 2002 Sexually dimorphic development of mouse primordial germ cells: switching from oogenesis to spermatogenesis. Development 129:1155–1164

    Bullejos M, Koopman P 2001 Spatially dynamic expression of Sry in mouse genital ridges. Dev Dyn 221:201–205

    Sekido R, Bar I, Narvaez V, Penny G, Lovell-Badge R 2004 SOX9 is up-regulated by the transient expression of SRY specifically in Sertoli cell precursors. Dev Biol 274:271–279

    Vidal VP, Chaboissier MC, de Rooij DG, Schedl A 2001 Sox9 induces testis development in XX transgenic mice. Nat Genet 28:216–217

    Foster JW, Dominguez-Steglich MA, Guioli S, Kowk G, Weller PA, Stevanovic M, Weissenbach J, Mansour S, Young ID, Goodfellow PN, Brook JD, Schafer AJ 1994 Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature 372:525–530

    Wagner T, Wirth J, Meyer J, Zabel B, Held M, Zimmer J, Pasantes J, Bricarelli FD, Keutel J, Hustert E, Wolf U, Tommerup N, Schempp W, Scherer G 1994 Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell 79:1111–1120

    Bi W, Huang W, Whitworth DJ, Deng JM, Zhang Z, Behringer RR, de Crombrugghe B 2001 Haploinsufficiency of Sox9 results in defective cartilage primordia and premature skeletal mineralization. Proc Natl Acad Sci USA 98:6698–6703

    Albrecht KH, Young M, Washburn LL, Eicher EM 2003 Sry expression level and protein isoform differences play a role in abnormal testis development in C57BL/6J mice carrying certain Sry alleles. Genetics 164:277–288

    Palmer SJ, Burgoyne PS 1991 The Mus musculus domesticus Tdy allele acts later than the Mus musculus musculus Tdy allele: a basis for XY sex-reversal in C57BL/6-YPOS mice. Development 113:709–714

    Schepers G, Wilson M, Wilhelm D, Koopman P 2003 SOX8 is expressed during testis differentiation in mice and synergizes with SF1 to activate the Amh promoter in vitro. J Biol Chem 278:28101–28108

    Tevosian SG, Albrecht KH, Crispino JD, Fujiwara Y, Eicher EM, Orkin SH 2002 Gonadal differentiation, sex determination and normal Sry expression in mice require direct interaction between transcription partners GATA4 and FOG2. Development 129:4627–4634

    Nef S, Verma-Kurvari S, Merenmies J, Vassalli JD, Efstratiadis A, Accili D, Parada LF 2003 Testis determination requires insulin receptor family function in mice. Nature 426:291–295

    Allard S, Adin P, Gouedard L, di Clemente N, Josso N, Orgebin-Crist MC, Picard JY, Xavier F 2000 Molecular mechanisms of hormone-mediated Mullerian duct regression: involvement of ?-catenin. Development 127:3349–3360

    Shen WH, Moore CC, Ikeda Y, Parker KL, Ingraham HA 1994 Nuclear receptor steroidogenic factor 1 regulates the mullerian inhibiting substance gene: a link to the sex determination cascade. Cell 77:651–661

    De Santa Barbara P, Bonneaud N, Boizet B, Desclozeaux M, Moniot B, Sudbeck P, Scherer G, Poulat F, Berta P 1998 Direct interaction of SRY-related protein SOX9 and steroidogenic factor 1 regulates transcription of the human anti-Mullerian hormone gene. Mol Cell Biol 18:6653–6665

    Viger RS, Mertineit C, Trasler JM, Nemer M 1998 Transcription factor GATA-4 is expressed in a sexually dimorphic pattern during mouse gonadal development and is a potent activator of the Mullerian inhibiting substance promoter. Development 125:2665–2675

    Vigier B, Watrin F, Magre S, Tran D, Josso N 1987 Purified bovine AMH induces a characteristic freemartin effect in fetal rat prospective ovaries exposed to it in vitro. Development 100:43–55

    Schmahl J, Kim Y, Colvin JS, Ornitz DM, Capel B 2004 Fgf9 induces proliferation and nuclear localization of FGFR2 in Sertoli precursors during male sex determination. Development 131:3627–3636

    Buehr M, Gu S, McLaren A 1993 Mesonephric contribution to testis differentiation in the fetal mouse. Development 117:273–281

    Tilmann C, Capel B 1999 Mesonephric cell migration induces testis cord formation and Sertoli cell differentiation in the mammalian gonad. Development 126:2883–2890

    Yao HH, DiNapoli L, Capel B 2003 Meiotic germ cells antagonize mesonephric cell migration and testis cord formation in mouse gonads. Development 130:5895–5902

    Britt KL, Drummond AE, Dyson M, Wreford NG, Jones ME, Simpson ER, Findlay JK 2001 The ovarian phenotype of the aromatase knockout (ArKO) mouse. J Steroid Biochem Mol Biol 79:181–185

    Brennan J, Tilmann C, Capel B 2003 Pdgfr- mediates testis cord organization and fetal Leydig cell development in the XY gonad. Genes Dev 17:800–810

    Kitamura K, Yanazawa M, Sugiyama N, Miura H, Iizuka-Kogo A, Kusaka M, Omichi K, Suzuki R, Kato-Fukui Y, Kamiirisa K, Matsuo M, Kamijo S, Kasahara M, Yoshioka H, Ogata T, Fukuda T, Kondo I, Kato M, Dobyns WB, Yokoyama M, Morohashi K 2002 Mutation of ARX causes abnormal development of forebrain and testes in mice and X-linked lissencephaly with abnormal genitalia in humans. Nat Genet 32:359–369

    Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC, Tonini G, Ferrante E, Chiumello G, McCabe ER, Fraccaro M, Zuffardi O, Camerino G 1994 A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet 7:497–501

    Swain A, Narvaez V, Burgoyne P, Camerino G, Lovell-Badge R 1998 Dax1 antagonizes Sry action in mammalian sex determination. Nature 391:761–767

    Meeks JJ, Weiss J, Jameson JL 2003 Dax1 is required for testis determination. Nat Genet 34:32–33

    McLaren A 2000 Germ and somatic cell lineages in the developing gonad. Mol Cell Endocrinol 163:3–9

    Matzuk MM, Lamb DJ 2002 Genetic dissection of mammalian fertility pathways. Nat Med 8(Suppl):S33–S40

    Menke DB, Koubova J, Page DC 2003 Sexual differentiation of germ cells in XX mouse gonads occurs in an anterior-to-posterior wave. Dev Biol 262:303–312

    McElreavey K, Vilain E, Abbas N, Herskowitz I, Fellous M 1993 A regulatory cascade hypothesis for mammalian sex determination: SRY represses a negative regulator of male development. Proc Natl Acad Sci USA 90:3368–3372

    Vainio S, Heikkila M, Kispert A, Chin N, McMahon AP 1999 Female development in mammals is regulated by Wnt-4 signalling. Nature 397:405–409

    Jeays-Ward K, Hoyle C, Brennan J, Dandonneau M, Alldus G, Capel B, Swain A 2003 Endothelial and steroidogenic cell migration are regulated by WNT4 in the developing mammalian gonad. Development 130:3663–3670

    Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ 2004 A WNT4 mutation associated with Mullerian-duct regression and virilization in a 46,XX woman. N Engl J Med 351:792–798

    Jordan BK, Mohammed M, Ching ST, Delot E, Chen XN, Dewing P, Swain A, Rao PN, Elejalde BR, Vilain E 2001 Up-regulation of WNT-4 signaling and dosage-sensitive sex reversal in humans. Am J Hum Genet 68:1102–1109

    Jordan BK, Shen JH, Olaso R, Ingraham HA, Vilain E 2003 Wnt4 overexpression disrupts normal testicular vasculature and inhibits testosterone synthesis by repressing steroidogenic factor 1/?-catenin synergy. Proc Natl Acad Sci USA 100:10866–10871

    Stark K, Vainio S, Vassileva G, McMahon AP 1994 Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature 372:679–683

    Brisken C, Heineman A, Chavarria T, Elenbaas B, Tan J, Dey SK, McMahon JA, McMahon AP, Weinberg RA 2000 Essential function of Wnt-4 in mammary gland development downstream of progesterone signaling. Genes Dev 14:650–654

    Treier M, Gleiberman AS, O’Connell SM, Szeto DP, McMahon JA, McMahon AP, Rosenfeld MG 1998 Multistep signaling requirements for pituitary organogenesis in vivo. Genes Dev 12:1691–1704

    Heikkila M, Peltoketo H, Leppaluoto J, Ilves M, Vuolteenaho O, Vainio S 2002 Wnt-4 deficiency alters mouse adrenal cortex function, reducing aldosterone production. Endocrinology 143:4358–4365

    Bishop CE, Whitworth DJ, Qin Y, Agoulnik AI, Agoulnik IU, Harrison WR, Behringer RR, Overbeek PA 2000 A transgenic insertion upstream of sox9 is associated with dominant XX sex reversal in the mouse. Nat Genet 26:490–494

    Gasca S, Canizares J, De Santa Barbara P, Mejean C, Poulat F, Berta P, Boizet-Bonhoure B 2002 A nuclear export signal within the high mobility group domain regulates the nucleocytoplasmic translocation of SOX9 during sexual determination. Proc Natl Acad Sci USA 99:11199–11204(Susan Y. Park and J. Larr)