Abstract
Alfred Jost's work in the 1940s laid the foundation of the current paradigm of sexual differentiation of reproductive tracts, which contends that testicular hormones drive the male patterning of reproductive tract system whereas the female phenotype arises by default. Once established, the sex-specific reproductive tracts undergo morphogenesis, giving rise to anatomically and functionally distinct tubular organs along the rostral–caudal axis. Impairment of sexual differentiation of reproductive tracts by genetic alteration and environmental exposure are the main causes of disorders of sex development, and infertility at adulthood. This review covers past and present work on sexual differentiation and morphogenesis of reproductive tracts, associated human disorders, and emerging technologies that have made impacts or could radically expand our knowledge in this field.
Keywords: androgens, androgen receptor, anti-Mullerian hormone, chromatin, developmental biology, developmental origins of health and disease, differentiation, environmental contaminants and toxicants, epididymis, epigenetics, female reproductive tract, fetal development, male reproductive tract, mechanisms of hormone action, Müllerian ducts, sex differentiation, testosterone, uterus, vagina, vas deferens, Wolffian duct
This review covers past and present work on sexual differentiation and morphogenesis of reproductive tracts, associated human disorders, and emerging technologies that have made impacts or could radically expand our knowledge in this field.
Introduction
Sexually dimorphic establishment of reproductive tracts is a critical step in developing reproductive tract organs anatomically and functionally distinct between male and female. Before sexual differentiation, an embryo possesses both female and male reproductive tract progenitors, which are also known as Müllerian and Wolffian ducts, respectively (Figure 1A). To establish sexual dimorphism of reproductive tract system, the embryo eliminates one of the two primitive ducts and maintains the other (Figure 1B). As a result, an embryo retains only one reproductive tract corresponding to its sex: Müllerian duct for the female and Wolffian duct for the male [1]. Müllerian ducts in the female eventually differentiate into the adult female reproductive tracts, which include oviduct, uterus, cervix, and the upper part of vagina (Figure 1C). These specialized components of the female reproductive tract create an environment that allows mating, fertilization, embryo transport, embryo implantation and development, and delivery of fetus. On the other hand, Wolffian ducts in the male develop into the adult male reproductive tract, which consists of epididymis, vas deferens, and seminal vesicle (Figure 1C). These male-specific organs facilitate the maturation, transport, storage, and delivery of the sperm. This review covers past and present work on sexual differentiation and morphogenesis of reproductive tracts, and related human developmental disorders. It also discusses unanswered questions and future opportunities brought forth by emerging technologies.
Figure 1.

Establishment of the sex-specific reproductive tract system. (A) Before sexual differentiation, the embryo possesses both primitive female and male reproductive tracts, which are known as Müllerian (represented in pink) and Wolffian ducts (in blue), respectively. (B) During sexual differentiation, the embryo eliminates one of the two primitive ducts and maintains the other. As a result, the XX embryo retains the Müllerian ducts and the XY embryo retains the Wolffian ducts. (C) Müllerian ducts in the XX embryo eventually differentiate into the adult female reproductive tracts, which include oviduct, uterus, cervix, and the upper part of vagina. On the other hand, Wolffian ducts in the XY embryo develop into the adult male reproductive tract, which consists of epididymis, vas deferens, and seminal vesicle.
Jost's paradigm: hormonal regulation of sexual differentiation
Jost's remarkable microsurgical experiments on rabbit embryos laid the foundation for our current knowledge of mammalian sexual differentiation [2, 3]. By removing the gonads of rabbit fetuses before the onset of sexual differentiation, Jost discovered that embryos without gonads, regardless of their genetic sex (XX or XY), acquired a female phenotype: regression of Wolffian ducts and maintenance of Müllerian ducts. He then grafted a fetal testis adjacent to the ovary of an XX embryo and found that the female pattern was reversed to the male pattern with the maintenance of Wolffian ducts and regression of Müllerian ducts. From these observations, Jost concluded that the female phenotypic development is a default process and masculinization of reproductive tract system is imposed by fetal testicular hormones.
At the time of Jost's experiment, testosterone was the only known hormone secreted by the testis for its ability to restore maleness in castrated animals and humans [4, 5]. By 1935, three research teams led by Adolf Butenandt, Kàroly Gyula, and Leopold Ruzicka, successfully synthesized the testicular hormone, ultimately named testosterone [6]. When Jost grafted a testosterone implant, instead of a fetal testis, to the XX embryo, Wolffian ducts were maintained but Müllerian ducts regression did not occur. This observation not only ascertained the function of testosterone on maintaining the Wolffian duct, but also implied that another testicular factor(s) must be responsible for inhibiting the survival of Müllerian ducts. Jost proposed the name “Müllerian inhibiting substance” or MIS for such factor, which is now known as anti-Müllerian hormone (AMH).
How do androgens promote the maintenance of Wolffian ducts?
In addition to Jost's experiments, the involvement of androgens on Wolffian duct maintenance was supported by various models of testicular feminization due to androgen insensitivity, including human [7], cattle [8], rat [9], and mouse [10]. Affected individuals were genetically XY that produced normal testicular androgens; however, they failed to be virilized. Genetic inheritance studies in mice revealed that testicular feminization was caused by an X-linked gene, which was named Tfm (testicular feminization mutation) [10]. After the cDNA of androgen receptor was cloned and mapped to the X chromosome in human and rat [11, 12], it became clear that the Tfm mouse was caused by a frameshift mutation in the androgen receptor gene, resulting in premature protein translation [13].
Androgen receptor (AR, or NR3C4) is a member of nuclear receptors with the conserved ligand binding domain and cysteine-rich DNA-binding domain [14]. Androgen receptors first appear in the Wolffian duct mesenchyme and later extend to both Wolffian duct epithelium and mesenchyme in mice and rat [15, 16]. However, in human embryos, androgen receptors seemed to be only expressed in Wolffian duct epithelium [17]. The transcriptional regulation of androgen receptor depends on androgen ligand activation. In the absence of androgens, androgen receptor is localized in the cytoplasm; upon binding to androgens, androgen receptor is translocated to the nucleus to regulate gene transcription [14]. The biosynthesis of androgens occurs in fetal Leydig cells [18]. The action of androgen receptor on Wolffian ducts must be imposed within a specific fetal programming window, which is E15.5–17.5 in rat and predicted to be approximately 8–12 weeks of gestation in human [18]. Inhibition of androgen receptor actions during this masculinization programming window disrupted stabilization and differentiation of Wolffian ducts [19–21].
The maintenance of Wolffian ducts is a product of androgen receptor action in the mesenchyme (Figure 2). Global knockout or loss-of-function mutation of androgen receptor resulted in failure to maintain Wolffian ducts in the XY mice [15, 22, 23]. However, ablation of androgen receptors only in the Wolffian duct epithelium had no impact on Wolffian duct maintenance, elongation, and coiling, supporting that action of androgen receptor occurs primarily in the Wolffian duct mesenchyme [15]. Mesenchymal cells of Wolffian duct also play instructing roles in Wolffian duct differentiation. When the epithelium from the upper Wolffian duct (future epididymis) was combined with the lower Wolffian duct mesenchyme (future seminal vesicle), the epithelium lost its epididymal identity and became seminal vesicle-like structures [24]. These results indicate that the fate and differentiation of Wolffian ducts are dictated by signals from the Wolffian duct mesenchyme.
Figure 2.
Mechanisms underlying the sex-specific establishment of the Wolffian duct. In the XX embryo, COUP-TFII in the Wolffian duct (or WD) mesenchyme suppresses Fgf expression and in the absence of FGF signaling and the Wolffian duct epithelium degenerates. On the other hand, in the XY embryo, testis-derived androgens activate androgen receptor (AR) and initiate the survival program via mesenchymal–epithelial crosstalk. The activated AR induces expression of growth factor EGF (epidermal growth factors) or other unknown factors in the mesenchyme. EGF binds its receptor EGFR in Wolffian duct epithelium, which presumably actives p-ERK and ETV4/5-mediated survival pathway in the epithelium. Additional AR action in the mesenchyme somehow antagonizes COUP-TFII’s inhibition of Fgf. As a result, FGF production is enhanced, which may promote Wolffian duct survival via the FGFR2/p-ERK pathway. Although EGFR and FGFR1 are expressed in the mesenchyme, it remains unclear about specific effects of EGF and FGF on mesenchymal cells during AR promoted Wolffian duct maintenance.
The mesenchymal androgen receptors control differentiation and survival of the Wolffian duct epithelium through induction of paracrine factors such as epidermal growth factor (EGF) (Figure 2). Androgen antagonist treatment to the XY mouse embryo decreased Egf expression [25], whereas androgen treatment to the XX embryos increased Egf and EGFR expression in the Wolffian duct [25, 26]. Additionally, in the absence of androgen, EGF treatment exerted androgenic effects on Wolffian duct maintenance and differentiation in organ culture and in vivo conditions [27]. Blocking EGF and EGFR action by their specific antibodies prevented androgen-induced effects on Wolffian ducts in organ culture [26, 27]. These results suggested that EGF/EGFR could be a downstream effector of androgen actions (Figure 2). Compound mutations in Egfr led to male infertility in mice but it is unknown whether Wolffian duct derivative tissues were abnormal in this mutant mouse [28]. In addition, Egf knockout mice had no abnormal phenotypes [29], suggesting that the EGF/EGFR pathway may be compensated by other growth factor signaling or alternatively dispensable for Wolffian duct development in vivo.
Fibroblast growth factor (FGF) is another candidate responsible for mediating mesenchymal AR actions (Figure 2). FGF7 and FGF10 are the major FGF ligands expressed in Wolffian duct mesenchyme [30–33]. Additions of FGF7, FGF10, or both were able to maintain Wolffian duct in the absence of testes in organ culture [32]. Among four receptors for FGFs in mammal, Fgfr2 is the major FGF receptor in Wolffian duct and its ablation led to caudal Wolffian duct regression [33]. While these observations implicate the involvement of FGF signaling in Wolffian duct maintenance (Figure 2), the connection between FGFs and androgen signaling remains to be determined.
Besides FGFs, growth hormone (GH), insulin-like growth factor (IGF1), and transformation growth factor beta 2 (TGFβ2) were implicated in Wolffian duct maintenance based on organ culture or ex vivo experiments. Both GH and its downstream mediator IGF1 were able to promote the maintenance of Wolffian ducts in the organ culture condition [34]. Igf1 knockout mice displayed underdevelopment of Wolffian duct derivative tissues including epididymis, seminal vesicle, and prostate. However, production of androgen, the driving hormone for Wolffian duct tissue development, was also reduced in Igf1 knockout males [35]. Therefore, it is unclear whether these hypoplastic Wolffian duct phenotypes in Igf1 knockout mice are the results of local IGF1 deficiency, androgen deficiency, or a combination of both. In the case of Tgfβ2, only one out of five examined Tgfβ2 knockout male mice had testis hypoplasia and vas deferens dysgenesis [36]. Further investigations are needed to affirm the physiological importance of these growth factors in Wolffian duct maintenance.
Although the maintenance of Wolffian duct during sexual differentiation is regulated through the action of mesenchyme, its survival prior to sexual differentiation depends on epithelial transcription factors, including homeobox gene Emx2 and paired box gene Pax2. In the absence of Emx2, although the Wolffian duct was initially formed, its structure degenerated in both sexes before the onset of sexual differentiation [37]. Likewise, loss of Pax2 caused degeneration of Wolffian duct and Müllerian ducts before the initiation of sexual differentiation despite normal-looking gonads, indicating an autonomous function of PAX2 in maintaining Wolffian ducts (as well as Müllerian ducts) [38]. These results implicate that maintenance of Wolffian duct epithelium is initially controlled by its own transcriptional programs until the onset of sexual differentiation, when androgen receptor signaling in the mesenchyme kicks in.
How does anti-Müllerian hormone induce the regression of Müllerian ducts?
Anti-Müllerian hormone belongs to the transforming growth factor beta (TGFβ) gene family with a C-terminal domain highly conserved with human TGFβ [39]. AMH is secreted specifically by Sertoli cells in both fetal and adult testes and granulosa cells of postnatal ovaries [40]. Purified AMH from incubation media of bovine fetal testicular tissue caused the elimination of Müllerian ducts in fetal XX rat mesonephros in culture [41, 42]. AMH cDNA and AMH gene in bovine and human were later cloned [39, 43]. Transfection of human AMH gene in Chinese hamster ovary cells led to AMH protein production, which induced Müllerian duct regression in organ culture assay [39]. Transgenic XX mouse embryos that express ectopic human AMH have no Müllerian duct derivative tissues [44], whereas Amh deficient XY mice retained female reproductive tract organs [45]. In humans, mutations in AMH are associated with persistent Müllerian duct syndrome in XY individuals [46]. The collective work unequivocally demonstrates the governing role of AMH in inducing Müllerian duct regression.
Being a member of the TGFβ gene family, anti-Müllerian hormone signals via heterotetrameric complexes of type I and type II kinase receptors in which the type II receptor phosphorylates and activates the type I receptor upon binding of the ligand [47]. The signaling is then transduced to the nucleus by members of the receptor-activated (R)-Smad family, which form trimeric complexes with the common mediator SMAD4 to regulate specific gene expression (Figure 3) [47]. The cDNA of type II receptor for AMH (Amhr2) was cloned from Sertoli cell, fetal ovaries, and fetal rat reproductive tracts [48–50]. The expression of the type II receptor gene is localized to the Müllerian duct mesenchyme during embryogenesis, and Sertoli cells and granulosa cells in fetal and adult testes and ovaries [48–51]. Expression of Amhr2 is directly controlled by the transcription factor Wilm's tumor or WT1 in the mesenchyme [52] (Figure 3). The functional significance of AMHR2 in transducing AMH signal was validated by the genetic mouse model: Amhr2 knockout XY mice exhibited Müllerian duct maintenance phenotype indistinguishable from Amh knockout and Amh/Amhr2 double knockout male mice [53]. These observations support that the pathway activated by AMH consists of only the ligand AMH and one type II receptor AMHR2. This notion was corroborated by clinical findings that XY patients with AMH or AMHR2 gene mutations caused the similar phenotypes of persistent Müllerian ducts [54].
Figure 3.
Mechanisms underlying sex-specific establishment of the Müllerian duct. In both sexes, Müllerian duct (or MD) mesenchyme expresses AMHR2, which is regulated by Müllerian duct epithelium-derived WNT7A and mesenchymal transcriptional factor WT1. It is unknown which specific FZ receptor (s) in the mesenchyme mediates WNT7A signaling to control Amhr2 expression. In the XX embryo, due to a lack of AMH, AMHR2-mediated signaling in Müllerian duct regression is not activated and as a result, Müllerian duct epithelium survives. It is unclear whether any mesenchyme-derived survival factor(s) are present to promote Müllerian duct maintenance. On the other hand, in the XY embryo, testis-derived AMH engages with AMHR2 and ALK2/3. Upon binding of AMH, AMHRII phosphorylates and activates ALK2/3. The signaling is then transduced to the nucleus by members of the receptor-activated (R)-Smad family (SMAD1/5/8), which form trimeric complexes with the common mediator SMAD4 to regulate specific gene expression, including WNT ligands Wnt4 and Wnt5a, WNT inhibitors such as Wif1, the transcriptional factor Osx, and the matrix metalloproteinase Mmp2. By unknown mechanisms, these molecules induce apoptosis or epithelial–mesenchymal transition (EMT), two major processes in Müllerian duct regression. Additionally, in the mesenchyme, secreted WNT ligands induce CTNNB1 (β-catenin)-dependent signaling in the nucleus, where the stabilized β-catenin engages and/or regulates TCFs and LEF1. These β-catenin-activated transcriptional factors regulate Osx and other unknown factors that participate in the regression of Müllerian duct epithelium.
The type I receptors for AMH and downstream R-Smads play redundant roles in mediating AMH-induced Müllerian duct regression. Two putative type I receptors for AMH in reproductive tract differentiation were identified as Alk3/Bmpr1a and Alk2/Acvr1, which are specifically expressed in Müllerian duct mesenchyme (Figure 3) [55–57]. Alk2 and Alk3 ablation in Amhr2+ mesenchymal lineages led to Müllerian duct retention in 0 and 50% examined male mice, respectively. However, the 100% penetrant phenotype was observed in Alk2 and Alk3 double mutant males [57]. The action AMH on Müllerian duct regression was abolished only when all the three R-Smad effectors (SMAD1, SMAD5, or SMAD8) were ablated in the Müllerian duct mesenchyme [57]. Ablation of the R-Smad common mediator Smad4 in MD mesenchyme led to only partial Müllerian duct retention on one side or both sides in XY mice [58].
Besides R-Smad effectors, another putative downstream transcriptional effector of AMH signaling in inducing Müllerian duct regression is the zinc figure transcriptional factor Osterix (Osx/Sp7) (Figure 3) [59]. Osx/Sp7 is specifically expressed in Müllerian duct mesenchyme in XY embryos. Amhr2 ablation in XY mouse embryos abolished Osx/Sp7 expression while ectopic expression of human AMH in XX mouse embryos induced its expression in Müllerian duct mesenchyme, implicating Osx as the direct downstream effectors of AMHR2 signaling. Ablation of Osx in Müllerian duct mesenchyme only caused delayed Müllerian duct regression in XY embryos, suggesting that Osx mediates only a part of AMH downstream signaling in inducing Müllerian duct regression.
It remains unclear how the AMH/AMHR2 signaling in the mesenchyme promoted the demise of Müllerian duct epithelium. Matrix metalloproteinases (MMPs) are calcium-dependent zinc-containing endopeptidases, which were hypothesized to promote apoptosis in Müllerian epithelial cells either by cleaving substrates on the epithelial cells or by modifying survival or death-inducing factors derived from the mesenchyme [60]. Among MMPs, Mmp2 was of great interest. Mmp2 expression was upregulated in male Müllerian duct mesenchyme and abolished in the absence of AMH, indicating Mmp2 was a potential downstream target of AMH signaling. However, Mmp2 expression was not regulated by AMH downstream transcriptional factor Osx based on the observation that Osx ablation in Müllerian duct mesenchyme did not change Mmp2 expression [59]. In ex vivo organ culture conditions, mature MMP2 led to increased incidence of Müllerian duct regression in the XX mouse embryos while Mmp2 knockdown partially blocked Müllerian duct regression in XY mouse embryos [60]. However, Mmp2 knockout mice appeared overall normal [61], suggesting that other Mmp members might compensate for the loss of Mmp2.
Crosstalk between AMH/AMHR2 and WNT signaling in regulating the fate of Müllerian ducts
The regression and maintenance of Müllerian duct during sexual differentiation is also dependent on WNT signaling, which interact with the AMH/AMHR2 pathway. WNT signal transduction cascade includes a WNT ligand (at least 19 Wnt genes were found in most mammalian including human and mouse), a heterodimeric receptor complex consisting of a seven-transmembrane receptor Frizzled (Fz), and a LRP5/6 coreceptor protein [62]. The WNT ligand Wnt7a is specifically expressed in Müllerian duct epithelium and required for Amhr2 expression in the mesenchyme (Figure 3). Wnt7a knockout XY mice had persistent Müllerian ducts due to the loss of Amhr2 in Müllerian duct mesenchyme [63].
In addition to Wnt7a, multiple WNT ligands and WNT ligand inhibitors have been suggested to involve in Müllerian duct fate decisions (Figure 3). Wnt4 is a downstream factor of AMH signaling and specifically expressed in Müllerian duct mesenchyme of XY embryos but not of XX embryos [64]. Wnt5a is another ligand expressed in the mesenchyme of developing murine Müllerian duct [65]. However, loss of Wnt4 [64] or Wnt5a [66] in Müllerian duct mesenchyme had normal Müllerian duct regression in XY embryos. Wnt4 and Wnt5a double ablation in Müllerian duct mesenchyme caused endometrial gland development and causes partial Müllerian agenesis in female mice but it was not reported whether Müllerian duct was persistent in the XY mice [67].
Secreted WNT inhibitors could participate in regulating the fate of the Müllerian duct (Figure 3). A WNT inhibitory factor Wif1 was identified as an AMH-regulated gene in Müllerian duct mesenchyme [68]. Knockdown Wif1 in organ culture condition led to partial or complete Müllerian duct maintenance in XY [68]. The secreted frizzled-related proteins (SFRPs) are a family of soluble proteins that are structurally related to frizzled (Fz) proteins. Because of their homology with the WNT-binding domain on the Fz receptors, SFRPs are able to inhibit WNT signaling [69]. Sfrp1 was expressed in the mesenchyme of both Müllerian duct and Wolffian duct [70]; Sfrp2 and Sfrp5 exhibited sexually dimorphic expression in Müllerian duct prior to the onset of regression [71]. These expression patterns suggest their potential roles in sexual differentiation of Müllerian ducts. However, knockout Sfrp1 and Sfrp2 caused a slight delay in Müllerian duct regression [70] and ENU-induced null mutations in Sfrp2 and Sfrp5 did not cause abnormalities in reproductive tract development at all [71]. These observations indicate that these WNT inhibitors could play redundant roles in regulating WNT signaling in Müllerian duct regression (Figure 3).
While essential roles of WNT ligands in regulating the fate of Müllerian duct are well established, receptor(s) for these WNT ligands have not been identified (Figure 3). There are 10 mammalian Fz receptors for WNT binding. So far Fz1 was the only receptor detected in Müllerian duct tissues by both RT-PCR and in situ hybridization. Fz1 was specifically expressed in Müllerian duct epithelium and its mesenchyme [72]. However, Fz1 knockout males were fertile, which indicates that they may have normal Müllerian duct regression [73]. Fz9 expression pattern in mesonephros was not reported during sexual differentiation [74] and Fz9 knockout had no Müllerian duct-related phenotypes [75]. Fz10 is specifically expressed in Müllerian duct epithelium [76], and its loss does not lead to Müllerian duct phenotype in mice (MGI: 3604450). Fz receptors may play redundant roles in mediating WNT signaling in the Müllerian ducts. Other than the Fz receptors, the glycosylphosphatidylinositol-anchored protein RECK was suggested to be a selective WNT7a receptor, and its binding with G protein-coupled receptor GPR124 modulated WNT7A-induced signaling in rat brain [77]. Reck is expressed in mesonephric mesenchyme during sexual differentiation of reproductive tract in mouse based on GUDMAP online database [78, 79]. These observations suggested a potential role of Reck in mediating WNT7a signaling in Müllerian duct differentiation. Since Reck knockout mice died at E10.5 [77], conditional ablation of Reck in Müllerian duct mesenchyme using Amhr2-Cre could shed light onto its potential role in Müllerian duct regression.
Upon activating membrane receptors, WNT ligands induce three major intracellular downstream pathways: the canonical pathway induced by β-catenin stabilization and two noncanonical β-catenin independent pathways, the WNT/Ca2+ and planar cell polarity (PCP) pathways [80]. Involvement of noncanonical β-catenin independent pathways in Müllerian duct regression has not been reported. On the other hand, the function of the β-catenin-dependent pathway in Müllerian duct fate decisions has been well documented (Figure 3). In response to the canonical WNT ligands, ligand-activated Fz receptors phosphorylate LRP, which breaks down the destruction complex and stabilizes β-catenin. The stabilized β-catenin accumulates and enters the nucleus where it engages DNA-bound TCF/LEF family transcription factors to drive target gene expression (Figure 3) [62]. Ablation of β-catenin in Müllerian duct mesenchyme led to Müllerian duct maintenance in XY mouse embryos [64]. In the absence of β-catenin, expression of the WNT target molecule, LEF1, was abolished in Müllerian duct mesenchyme [64]. Therefore, in the normal condition, β-catenin could regulate and engage LEF1 for target gene expression to induce Müllerian duct regression (Figure 3) [64]. However, Lef1 null XY mice had normal Müllerian duct regression [81], suggesting that action of β-catenin may be mediated by other TCF/LEF family transcriptional factors (Figure 3). Counterintuitively, when β-catenin was constitutively activated in Müllerian duct mesenchyme, focal maintenance of Müllerian duct in XY mouse embryos was observed [82]. These observations indicate that a properly tuned activity of β-catenin is critical for Müllerian duct regression.
The WNT/β-catenin and AMH/AMHR2 pathways interact and converge on β-catenin intracellularly in regulating the Müllerian duct fate. Nuclear staining of β-catenin was promoted by AMH signaling in Müllerian duct mesenchyme of XY embryos [83]. In the absence of AMH downstream effectors Smads, β-catenin expression was locally reduced (Figure 3) [58]. These observations indicate that AMH/AMHR2/SMAD modulates β-catenin expression and/or stabilization [64]. On the other hand, specific ablation of β-catenin in the Müllerian duct mesenchyme reduced expression of the AMH/AMHR2 downstream effector Osx [59]. However, β-catenin ablation did not affect the expression of Amhr2 and the AMH downstream signaling Wnt4 [64]. Another AMH downstream signaling Mmp2 [60] is a well-known target of Wnt/β-catenin signaling in cancer cells [84] but whether Wnt/β-catenin signaling regulates Mmp2 in Müllerian duct regression is unknown. Therefore, β-catenin as the core player of the canonical WNT pathway modulates partial AMH/AMHR2 signaling pathway for Müllerian duct regression (Figure 3).
Revisiting Jost's paradigm: hormone-independent sexual dimorphism of urogenital traits
While Jost's paradigm holds true in both animal studies and clinical cases, emerging evidence prompts some reconsideration of this dogma. The Jost paradigm contends that the female pattern of reproductive system occurs by default as the presence of Müllerian duct and absence of Wolffian duct being the product of a lack of fetal testes and their hormone-product androgens and AMH. However, the discovery of an unexpected role of a mesenchymal transcription factor COUP-TFII (chicken ovalbumin upstream promoter transcription factor II or NR2F2) added new twists and insights into this process. When Coup-tfII was inactivated in the Wolffian duct mesenchyme in the XX embryos, Wolffian ducts were maintained despite the absence of androgen and its action [32]. As a consequence of Coup-tfII inactivation, Fgf 7 and Fgf10 expression was increased along with the activation of its receptor (FGFR2) and downstream pERK-mediated cell survival signaling in the Wolffian duct epithelium, leading to the maintenance of Wolffian duct. These findings imply that in the androgen-free female embryo, the Coup-tfII/Fgf/pERK pathway inhibits Wolffian duct survival (Figure 2) and COUP-TFII serves as a “Wolffian inhibiting factor.” Being a member of the orphan nuclear receptor family, COUP-TFII is not known to be activated by endogenous ligands. It was reported that COUP-TFII can be activated by retinoic acid (RA) in multiple cell lines [85]. During sexual differentiation, RA is produced at high levels in the mesonephros where the Wolffian ducts develop, suggesting that RA could potentially activate COUP-TFII [86].
Hormone-independent establishment of reproductive traits, similar to the maintenance of Wolffian ducts without androgen action in the Coup-tfII knockout XX mouse embryos, is observed in other occasions. For example, female spotted hyenas and elephants develop male-type external genitalia prior to gonadal differentiation, suggesting that the masculinization of external genitalia in these two species is not regulated by fetal testes [87]. In wallabies, sexual dimorphisms are already present before differentiation of the gonads, with the scrotum developing in males and the pouch and mammary glands in females. The sexually dimorphic development of these tissues is believed to be controlled by yet-to-identified gene(s) on the X chromosome [88]. These cases of hormone-independent sexual dimorphisms argue against the simplicity and generality of Jost's paradigm, which holds true in majority of cases but need to be refined in specific contexts.
Molecular pathways that direct rostral-caudal patterning of the reproductive tract
Once sexual dimorphism of reproductive tract system is established, the stabilized Wolffian duct and Müllerian duct differentiate to morphologically and functionally distinct tubular organs in XY and XX embryos. From rostral to caudal regions, Müllerian ducts eventually differentiate into oviduct, uterus, cervix, and the upper part of vagina in XX (Figures 1 and 4); on the other hand, Wolffian ducts in XY develop into epididymis (consisting of four major regions: initial segment, caput, corpus, and cauda), vas deferens, and seminal vesicle (Figures 1 and 4). Despite the fact that maintenance and regression of two reproductive tracts is governed by testicular hormones, rostral-caudal patterning of two reproductive tracts is less dependent on gonadal hormones and their signaling pathways, and becomes predominantly regulated by epithelial–mesenchymal interactions.
Figure 4.
Putative morphogenetic factors and their expression in rostral-caudal patterning of the reproductive tract. During rostral-caudal patterning, the male reproductive tract differentiates into epididymis (consisting of four major regions: initial segment, caput, corpus, and cauda), vas deferens, and seminal vesicle (A); the female reproductive tract gives rise to oviduct, uterus, cervix, and the upper vagina (B). Mesenchymal–epithelial interaction is the core mechanism in establishing rostral-caudal patterning of the two reproductive tracts. The model depicts expression localization of mesenchymal factors (marked in purple rectangles) and epithelial factors (in green rectangles) along the rostral-caudal axis. Inverted triangle indicates gradient expression from rostral to caudal region. Hox, homeobox; Inhba, inhibin beta A; Wnt, wingless/integrated; Bmp, bone morphogenetic protein; Ar, androgen receptor; Pkd1, polycystic kidney disease 1; components in RTK signaling, receptor tyrosine kinase) [Ros1 (Ros proto-oncogene 1, also known as cRos, an orphan receptor tyrosine kinase), Shp1 (Src homology region 2 domain-containing phosphatase-1), ERK (also known as MAPK, mitogen-activated protein kinase), Dusp6 (dual specificity phosphatase 6) and Pten (Phosphatase and tensin homolog)], p63, tumor protein p63; RA, retinoic acid; PCP proteins, planar cell polarity; Vangl2, Vang-like protein 2; and Celsr1, cadherin EGF LAG seven-pass G-type receptor 1.
Epithelial–mesenchymal interaction is the fundamental mechanism in patterning tubular structures [89]. During organogenesis, regional specification of the epithelium depends upon the identity of the adjacent mesenchyme. The critical role of the mesenchyme in the epithelial differentiation of male and female reproductive tracts has been established by tissues recombination studies [24, 90, 91], where mesenchyme of a specific origin and epithelium of a different origin were recombined and cultured to determine whether the fate of the epithelium can be altered. For example, when cultured with uterine mesenchyme, vaginal epithelium differentiates into uterine epithelium. In the reciprocal tissue recombinants, vaginal mesenchyme transformed the uterine epithelium into a stratified squamous vaginal epithelium [90]. Similar recombination experiments using Wolffian duct tissues have been conducted wherein the upper Wolffian duct (future epididymis) was combined with the lower Wolffian duct mesenchyme (future seminal vesicle). The epithelium of the upper Wolffian duct lost its epididymal identity and became seminal vesicle-like structures in response to induction by lower Wolffian duct mesenchyme [24]. Therefore, during the reproductive tract patterning, mesenchyme is probably differentiated first and then the preceding specified mesenchyme induces epithelial region-specific phenotype. Despite the critical roles of the mesenchyme in inducing epithelial differentiation, the interaction between the mesenchyme and epithelium is not a one-way street. Epithelium-derived signaling molecules are important for proper mesenchymal differentiation as well (see the details in next two sections). Although epithelial–mesenchymal interaction is the common mechanism underlying patterning of two reproductive tracts, the detailed pathways in patterning the two tubular organs are not exactly the same.
Patterning of the male reproductive tract
Mesenchymal differentiation by Homeobox genes along rostro-caudal axis is at the core of Wolffian duct patterning (Figure 4A). Hoxa9 and Hoxd9 are expressed in the epididymis and vas deferens (Figure 4A) [92]. Approximately half of the Hoxa9/Hoxd9 double knockout mice were fertile in these mice [93], suggesting that Wolffian duct development was probably normal and loss of Hoxa9 and Hoxd9 might be compensated by other Hox genes. The Hoxa10 expression is mainly in the mesenchymal compartments of caudal epididymis throughout the distal male reproductive tract during morphogenesis (Figure 4A) [94]. Hoxa10 mutant males had anterior transformation of the cauda epididymis and the proximal vas deferens, diminished stromal clefting of the seminal vesicles, and decreased size and branching of the coagulating gland [94, 95]. Hoxa11 is expressed in the mesenchyme of fetal Wolffian duct and adult vas deferens (Figure 4A). Either individual Hoxa11 knockout or Hoxa11 and Hoxd11 (a paralog to Hoxa11) double knockout caused malformations of vas deferens that resembled an epididymis [96, 97]. Hoxa13 and Hoxd13 are expressed in the distal part of Wolffian duct tissues (Figure 4A) [98]. Disruption of Hoxd13 alone led to hypoplastic seminal vesicles, which become more severe in Hoxa13+/– and Hoxd13–/– compound mutant mice [99]. In addition to Hox9–13, comparing gene transcriptional profiles of different Wolffian duct parts within the morphogenesis window has identified additional region-specific Hox transcripts, such as Hoxc4 and Hoxc9 in epididymis [100]. Specific functions of these newly identified Hox genes in Wolffian duct differentiation remains to be studied. The importance of Hox genes in determining the rostral-cauda patterning of the Wolffian duct is well recognized; nevertheless, it is not known how these Hox transcription factors induce morphology and phenotype variation in different parts of the Wolffian duct.
The specified mesenchyme provides signaling inputs via paracrine signaling to instruct epithelial differentiation in the male reproductive tract. Inhibin beta A (Inhba), a component of inhibins and activins (members of TGFβ superfamily ligand), is highly expressed in the mesenchyme of the anterior Wolffian duct and diminishes posteriorly before the onset of Wolffian duct morphogenesis (Figure 4A). Inhba mutant mice failed to develop epididymal coiling due to a dramatic decrease in epithelial proliferation [101], indicating its critical function in inducing the highly convoluted epididymal structure. In addition to inhibin beta A, BMPs and WNTs are other putative mesenchymal factors that control epididymal patterning: the epididymal epithelium failed to coil in the absence of Pkd1, a large structural membrane spanning glycoprotein involved BMP/Tgfβ signaling transduction [102], or β-catenin [103](Figure 4A).
Although epithelial differentiation is regulated by paracrine signaling from the mesenchyme, signaling pathways innated to the epithelium are also necessary to achieve its proper and complete differentiation. Androgen receptor in the Wolffian duct epithelium controls differentiation of basal cells in the epididymal epithelium by regulating p63, a driver for basal cell differentiation (Figure 4A) [15]. Although androgen receptor is expressed in other parts of Wolffian duct derivative tissues, androgen receptor regulation of basal cell differentiation is specific to the epididymis. The specific function of androgen receptor in the epididymis could be achieved by the pioneering factor AP-2α (activating enhancer binding protein 2α), which guides AR to specific genomic loci for epididymal gene regulation [104]. However, AP-2α knockout mice do not seem to have any reproductive defects (MGI: 104671), indicating that AP-2α can be compensated by other pioneering factor(s) or is dispensable for androgen receptor action in inducing basal cell differentiation in the epididymis.
Additionally, receptor tyrosine kinase (RTK) signaling in the epithelium also regulates establishment and/or maintenance of epithelial identity in the epididymis, espeically the initial segment. Multiple receptor-associated components in RTK pathways are highly expressed in the epithelium of initial segment of the mouse epididymis (Figure 4A), including an orphan receptor tyrosine kinase Ros1 (also known as c-Ros) [105], its negative regulator Shp1 [106], and fibroblast growth factor receptor substrate 2 (Frs2) [107]. Both Ros1 mutation and a naturally occuring mutation in Shp1 caused the complete loss of the regional characteristics of the initial segment. The proximal epididymal epithelium lacking Frs2 gave rise to abnormal shape of promixal epididymis with a range of variety in a subgroup of mice (∼10% examined animals) [107]. In addition, downstream pathways of RTK, such as MAPK/ERK and PIP3/AKT pathways, exhibited higher activity in the initial segment, including p-ERK1/2 and Src (components in ERK pathways), Dusp6 (a negative regulator of MAPK/ERK), and Pten (a negative regulator of PIP/AKT signaling) [108]. p-ERK1/2 and Dusp6 were critical for the proliferation of the initial segment and the caput & corpus regions, respectively [108]. Pten is required to maintain the epithelial fate in the initial segement because postnatal ablation of Pten (from postnatal P17 onward) induced dedifferentiation of the initial segment [109]. Even though multiple RTK pathways are well characeterized in regulating differentiaton or/and maintenance of the initial segment, specific factor(s) that engage and activate tyrosine kinase receptors has yet to be discovered.
Patterning the female reproductive tract
Similar to that in the male, region-specific Hox genes determine mesenchymal differentiation during the female reproductive tract patterning as well. Comparable to Hox gene expression along the anterior-posterior axis of the male reproductive tract, expression of Hoxa9, Hoxa10, Hoxa11, and Hoxa13 in the female reproductive tract is restricted to the oviduct, the uterus, the posterior uterus and cervix, and the cervix and upper vagina, respectively (Figure 4B) [110]. Genetic polymorphisms in HOXA9 have been linked to Müllerian duct abnormalities in humans [111] even though Hoxa9 knockout mice did not have any defects in urogenital organs (MGI: 96180). Hoxa10 and Hoxa11 are expressed in the mesenchyme of Müllerian ducts region that eventually becomes uterus. Both Hoxa10 and Hoxa11 null mutation led to partial morphological shift of the uterus towards the appearance of the rostral oviducts [95, 96]. Absence of Hoxa13 led to agenesis of cauda Müllerian ducts [99, 112]. Hoxa13+/− and Hoxd13−/- (a paralog to Hoxa13) compound mutations led to homeotic transformation of cervix to uterus [99].
Besides Hox genes, RA signaling regulates mesenchymal differentiation of Müllerian duct. Retinoic acid signaling is transduced by the two families of RA nuclear receptors (RARs and RXRs) during development. High expression of RA producing enzymes and transactivation activity was detected in the proximal Müllerian ducts that develop into the uterus (Figure 4B) [113]. In organ culture of undifferentiated Müllerian ducts, RA treatment induced uterus-specific Hox genes and uterine stromal differentiation, whereas inhibition of RA receptor signaling induced vaginal stromal differentiation [113]. These in vitro culture studies indicate that RA signaling determines the border between uterine and vaginal mesenchyme differentiation [113]. Ablation of RA receptors in mice would help us further elucidate significant roles of RA signaling in Müllerian duct mesenchymal differentiation in vivo. However, RAR mutations and ablation of RAR with RXR in mice led to differential Müllerian duct agenesis [114, 115], preventing the investigation of RA signaling in Müllerian duct patterning in vivo.
Mesenchymal regulation of epithelial differentiation is mediated in part by WNT signaling. Mesenchyme-derived Wnt5a is required for patterning of multiple regions in the female reproductive tract (Figure 4B). Wnt5a null mutation caused short and coiled uterus with no glands and the loss of defined cervical/vaginal structures [66]. Another Wnt ligand of note is Wnt4, which is expressed in uterine mesenchyme at birth prior to Müllerian duct morphogenesis (Figure 4B) [65]. Postnatal ablation of uterine Wnt4 led to transformation of the columnar epithelium to a stratified epithelial layer, a characteristic of the cervix and vagina [116], suggesting that mesenchyme-derived Wnt4 regulates uterine epithelial fate.
While it is well known that mesenchyme-derived WNT signaling plays significant roles in regulating epithelial differentiation, how the WNT signaling exerts their actions in the epithelium has yet to be fully understood. WNT ligands signal through three major well-characterized intracellular pathways: a canonical β-catenin-dependent pathway and two non-canonical β-catenin independent pathways, the WNT/Ca2+ and PCP pathways [80]. It is noteworthy that PCP proteins are expressed in Müllerian duct epithelium during morphogenesis and PCP gene mutations led to impaired Müllerian duct patterning (Figure 4B) [117, 118]. The WNT/PCP pathway regulates cell motility and polarity through cytoskeletal changes [119]. VANGL2 is an archetypal WNT/PCP protein, and a point mutation in Vangl2 led to dislocalized VANGL2 and in turn altered epithelial polarity. The female reproductive tract with mutant Vangl2 had septate vaginas, uncoiled oviduct, hyperplastic myometrium, and hypoplastic stromal mesenchyme in the uterus [117]. Besides, loss of a VANGL2-associated PCP gene Celsr1 (cadherin EGF LAG seven-pass G-type receptor) resulted in discontinuities of oviduct and uterus, aberrant epithelial folding, shape and arrangement in the oviduct [118]. Undoubtedly, the noncanonical WNT/PCP pathway is critical for appropriate establishment of epithelial polarity in Müllerian duct patterning. Phenotypes of PCP mutant mice are dissimilar to Wnt4 or Wnt5a null mice, suggesting that mesenchymal WNT signaling via WNT4 or WNT5a is probably not transduced through the PCP intracellular pathway in the epithelium.
Besides WNT, FGF7/10, BMP4, and Activin A from the mesenchyme also regulate epithelial patterning, particularly epithelial stratification in the cervix and vagina (Figure 4B). These mesenchymal factors signal through their respective receptors and intracellular pathways FGF/MAPK, BMP/Smad4, and Activin A/RUNX1, respectively, to cooperatively induce cervical and vaginal stratification [120]. Disruption in either of these pathways converts vaginal to uterine epithelial cell fate by inducing P63 expression [120]. P63 is the transcriptional factor that induces the epithelial stratification program [121]. Cervical and vaginal epithelia lacking p63 differentiated into columnar, uterine-like epithelium [122],
Although epithelial differentiation is regulated by mesenchymal signals, interaction between these compartments in the female reproductive tract is a two-way street. The epithelium of Müllerian ducts expresses Wnt7a, which becomes more restricted to oviductal and uterine parts before morphogenesis (Figure 4B) [65]. Wnt7a null mutation in mice caused posterior transformation of oviduct to uterus and uterus to vagina [123], implicating that epithelium secreted Wnt7a is critical for mesenchymal differentiation of the oviduct and uterus. Absence of β-catenin in the mesenchyme of the female reproductive tract do not recapitulate Wnt7a phenotype [72, 124], indicating that the action of WNT7A on mesenchymal differentiation is mediated by an unknown β-catenin independent mechanism.
Disorders of sexual differentiation of reproductive tracts in humans
Sexually dimorphic establishment of reproductive tracts can be perturbed by genetic alteration and environmental exposure, leading to human disorders that feature absence and persistence of reproductive tracts. Phenotypes of these human disorders are consistent with Jost's paradigm.
Androgen insensitivity syndrome (AIS) is a disorder of sexual differentiation due to inactivating mutations in androgen receptor gene that confer a spectrum of androgen resistance in the XY patients. AIS is clinically subclassified into three categories: (1) complete form (CAIS) that exhibits normal female external genitalia and a complete absence of Wolffian duct-derived organs, (2) mild form (MAIS), which present normal male external genitalia but may show gynecomastia and impaired spermatogenesis, and (3) partial form (PAIS) that presents phenotypes between CAIS and MAIS (please see reviews [125–127]). Fetal exposure to endocrine disrupting chemicals in humans is associated with phenotypes similar to androgen insensitivity syndromes [128, 129].
Persistent Müllerian duct syndrome (PMDS) is defined as the presence of ectopic female reproductive tract organs, including uterine and fallopian duct tissue, in XY patients that are normally virilized [46]. In these XY patients, undescended testes and abnormalities in male excretory ducts are frequent, and infertility is the most common complication of PMDS. Mutations in AMH/AMHRII were found in most PMDS patients [46]. In patients where no mutation in AMH/AMHRII were found, mutations in distal promoter or intron of AMH/AMHRII, or other alterations in genetic pathways implicated in Müllerian duct development, were thought to be responsible for these idiopathic PMDS. Prenatal exposure to a synthetic estrogen diethylstilbestrol increased the incidence of presence of Müllerian duct remnants in human [130], suggesting that estrogenic environmental chemicals are a major risk factor that contribute to persistent Müllerian duct syndrome. PMDS was also associated with prostate cancers in two clinical cases [131, 132]. In mice, residual Müllerian duct mesenchyme contributes to prostate tissues, and Lkb1 deletion in Müllerian duct mesenchyme led to the formation of benign prostatic hyperplasia [133]. This finding in mice raises the possibility that prostate tumors in reported PMDS patients may be of Müllerian duct mesenchyme origin.
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a disorder that affects 1 in 4500 females, and is characterized by the absence of Müllerian duct structures [134]. It can occur independently or coupled with various abnormalities. Whole-exome sequencing indicated that loss-of-function variants in OR4M2 (olfactory receptor, family 4, subfamily M, member 2) and PDE11A (phosphodiesterase 11A), nonsynonymous variants in LRP10 (LDL receptor related protein 10) and DOCK4 (Dedicator of cytokinesis 4), as well as deletions at 16p11.2, 15q11.2, 19q13.31, 1p36.21, and 1q44 were associated with MRKH syndromes [135, 136]. Exposure of rat fetuses to phthalate can cause uterine agenesis, a characteristic phenotype in MRKH syndrome [137]. However, in humans, exact contributions of nongenetic factors to the pathogenesis of MRKH are unclear [138].
Gartner's ducts are the vestigial remnants of Wolffian ducts in the vaginal regions of XX patients [139]. The presence of Gartner ducts is usually clinically asymptomatic, but they can grow large enough to form symptomatic cysts. The Gartner's duct cyst can mimic pelvic organ prolapse because both syndromes may exhibit a cystic mass posterior/anterior to vagina [140, 141]. Malignant transformation of Gartner cysts is very rare but a case was reported in the literature, which was treated successfully with surgical excision and radiation therapy [142]. In utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin at the time of sexual differentiation led to persistent Wolffian duct remnants in the rat vagina [143]. However, the association of dioxin exposure with Wolffian duct maintenance has not been reported in humans.
Remaining questions and future opportunities presented by emerging technologies
Visualizing the tridimensional cellular and morphological processes during sexual differentiation and morphogenesis of reproductive tracts
Generating a precise cellular and molecular cartography of reproductive tract system is critical for the understanding of the mechanisms of its organogenesis. Tridimensional imaging techniques will help uncover novel cellular and morphological events that cannot be visualized in histological and 2D microscopic analyses. For example, combination of whole mount immunostaining, solvent-based clearing, and light-sheet imaging has successfully built a 3D cellular map of the human development during the first trimester of gestation [144]. The 3D imaging of human reproductive tract system during sexual differentiation windows shed some interesting insights [144]. In contrast to Wolffian ducts that were vascularized in both sexes, the 3D imaging technique revealed sexually dimorphic vascularization surrounding the Müllerian ducts, which was unsheathed by vascular network in females but avascular in males [144]. This observation suggested that lack of vascularization may facilitate Müllerian duct regression in human males. Confocal imaging in combination with 3D analysis was also successfully applied to identify and quantify morphological changes in the luminal structure of murine uterus during embryo implantation, discovering a stereotypical reorientation of glandular ducts towards the site of implantation [145]. In addition to confocal imaging, optical coherence tomography is another powerful imaging technique, which penetrates more deeply into the sample to capture micrometer-resolution 2D or 3D images within live biological tissues. Given that this imaging method has successfully created high-resolution 3D in vivo imaging of mouse oviduct and visualize cellular events in oviducts [146–148], it can be applied to obtain live images of developing reproductive tract organs at fetal and postnatal stages. Hopefully, utilization of these 3D imaging approaches will create a molecular, cellular, and morphological atlas of reproductive tract organs, which will be of paramount importance to understanding its organogenesis in normal and pathological conditions.
Deciphering the molecular mechanisms underlying human reproductive tract differentiation
Although anatomical, histological, and microscopic analyses of human fetal tissues and the genetics of developmental disorders in reproductive tracts have helped us understand how human reproductive tracts form and develop [81, 149–151], knowledge about molecular mechanisms underlying this organogenesis in human is still limited. One major challenge is that we lack an in vitro system, which does not rely on materials from human fetuses, in order to model in vivo human organogenesis. Tridimensional organoid culture is a technique that can overcome this challenge. Organoids are progenitor cell-derived or stem cell-derived 3D structures that, on much smaller scales, re-create important aspects of the 3D anatomy and multicellular repertoire of their physiological counterparts and that can recapitulate basic tissue-level functions [152]. Endometrial and oviductal organoids deriving from adult mouse and human adult tissues have been successfully established [153–156]. In both species, continuous growth and differentiation of uterine and oviductal organoids depend on WNT and Notch signaling, respectively, suggesting conserved roles of these two pathways in female reproductive tract differentiation in mouse and human [153–156]. In addition to progenitor cells in adult tissues, organoid culture can derive from human-induced pluripotent stem cells (iPSCs). Based on results from studying in vivo mouse or human organ development, researchers have successfully directed differentiation of human iPSCs through intermediate mesoderm, coelomic epithelium into fallopian tube organoid [157], or uterine endometrial stromal fibroblasts [158]. By introducing specific signaling cues or alterations in the process of directed differentiation, we can investigate functional significances of these signaling factors underlying fetal human reproductive tract differentiation that otherwise cannot be studied due to tissue inaccessibility.
Determining the roles of epigenetic/chromatin-based mechanisms in sexual differentiation and morphogenesis of reproductive tracts
Signaling pathways and transcriptional regulation in reproductive tract development are well characterized (see the reviews [81, 151, 159]). It is now widely accepted that chromatin accessibility and its spatial 3D architecture play a crucial role in transcriptional regulation and subsequent cellular differentiation. Specialized packaging (or “closing”) of the chromatins sequesters inactive genomic regions while its “opening” exposes genomic active regions (promoter, enhancers and other regulatory elements) accessible to transcription machinery [160]. The spatial organization of chromosomes arrangement facilitates communication between genes and their regulatory elements, which can be located elsewhere along the chromosome [161]. Experimental and computational methods in chromatin biology, such as ATAC-seq for assessing chromatin accessibility, and chromosome conformation capture (3C) and 3C-derived methods for examining spatial chromatin arrangements, have provided a rich trove of information about promoter, enhancer, other regulatory elements, and their interactions in gene expression [161, 162]. Genome-wide chromatin structure changes has been suggested to involve in Müllerian duct differentiation in chicken, where chromatins of Müllerian ducts at different stages show different affinity with an antibody against chick newborn oviducts [163]. This observation indicates that chromatin changes may determine the developmental programming of accurate genomic expression and hormone responsiveness in the sexual differentiation tissues. The established techniques in chromatin biology will be powerful tools to test this notion. In addition to probe chromatin-based mechanisms in normal condition, another application of techniques for chromatin analysis is to understand chromatin organization alteration in pathological conditions. As discussed in the previous section, chromosome deletions are known to be associated with certain human disorders of reproductive tract development. While loss of genes within deleted regions can result in pathogenesis, it remains unclear whether the loss of noncoding chromosomal regions can disorganize chromatin arrangement and thus cause misregulation of expression of critical genes localized outside the deletion regions.
Categorizing the heterogeneous populations of cells in reproductive tract organs
Understanding cellular heterogeneity, especially in the mesenchyme and epithelium, will help us understand how mesenchymal–epithelial interactions along the rostral-caudal axis differ and drive regionalization. A powerful tool for dissecting heterogeneity in tissues is single cell RNA-seq (scRNA-seq), which permits transcriptomic analysis of individual cells. Analyzing postnatal mouse uterus by scRNA-seq uncovered a novel subpopulation of early progenitor in the epithelium [164] and chemokine (C-X-C motif) receptor 4; chemokine (C-X-C motif) ligand 12 signaling axis in mesenchymal–epithelial interaction for uterine gland formation [165]. Additionally, scRNA-seq analysis can provide a panorama of the entire cell types/subtypes in addition to epithelium and mesenchyme, which is essential to fully categorize the building blocks of reproductive tracts. Cell heterogeneities of human female reproductive tract have been revealed in a scRNA-seq analysis of human uterus, where 11 transcriptionally distinct clusters of epithelia, 6 clusters in stromal cells, 5 in endothelial cells, 2 in smooth muscle cells, 2 myofibroblasts, and 6 in immune cells have been found (https://doi.org/10.1101/267849). Single-cell transcriptomic analysis of fetal reproductive tract tissues will enable us to understand cellular heterogeneities and provide a rich resource for investigating signaling regulation network, especially in mesenchymal–epithelial interactions.
Establishing the roadmap of cell lineage specification during reproductive tract differentiation
A cell lineage is the developmental history of a differentiated cell as traced back to its progenitor from which it arises. Resolving lineage relationships between cells in the developing reproductive tracts, and between an early progenitor and its descendants, will illuminate mechanisms underlying normal development. A popular method for tracing lineage in genetically accessible model organisms such as mouse is the fate-mapping technique, which leverages the expression of recombinase enzymes in a cell-specific or tissue-specific manner to activate the expression of a reporter [166]. As a result, the cell/tissue and their progeny will be labeled with the reporter for tracking their fate as they differentiate. This approach has been used to investigate lineage differentiation in female reproductive tract formation in two studies. One study found that labeled secretory cells in the oviduct had the capacity to self-renew and gave rise to another oviductal cell type, ciliated cells, implicating critical roles of secretory cells in epithelial homoeostasis [167]. The other study discovered that Müllerian duct epithelial cells were partly derived from Wnt4+ lineage [168], suggesting that multiple lineages contribute to Müllerian duct cells. Noticeably, simultaneous lineage tracing of large-scale number of cells has been developed to depict lineage trees in zebrafish [169] and mouse [170]. The main principle is to utilize barcodes that consist of multiple CRISPR/CAS9 target sites to label cells. The barcodes will progressively and stably accumulate unique mutations over multiple cellular divisions and targeted sequencing analyses of the patterns of shared mutations will elucidate lineage relationships of analyzed cells. If inheritable cellular barcodes are transcribed, scRNA-seq can be used to simultaneously read the lineage barcodes and provide transcriptome information of single cells. Combination of computational analysis of lineage barcodes and single-cell RNA-seq has enable organization of the taxonomy of cell types into lineage tree in zebrafish [171–173]. If eventually applicable to mouse organism, this systematic and simultaneous assay of lineage relationship and cell types will provide unparalleled information on reproductive tract development.
Conclusions
Like a prologue in a play, Jost's classic experiments in the 1940s trailblaze the new frontier the field of sexual differentiation. Since then, significant insights into the establishment of sexually dimorphic reproductive tract system were gained from ex vivo experiments, genetic animal models, and human clinical cases. With the technological advancements and multidisciplinary collaborations, the field is poised to connect epigenetic mechanisms, molecular pathways, cellular events, and tridimensional morphological changes underlying the process of reproductive tract morphogenesis. The yielded knowledge will enable us to not only decipher the fundamental process of dimorphic establishment of reproductive tracts, but also provide insights into how defects and diseases originate from impaired fetal development.
Notes
Edited by Dr. Romana Nowak, PhD, University of Illinois Urbana-Champaign
Author Biographical
Humphrey H-C Yao, Senior Principal Investigator, and Fei Zhao, NIH Intramural Postdoctoral Fellow, in the Reproductive Developmental Biology Group at NIEHS/NIH. Dr. Humphrey H-C Yao received his doctoral degree at the University of Illinois in Urbana-Champaign in 1999 under the guidance of Janice Bahr, and then completed his postdoctoral training in Blanche Capel's lab at Duke University Medical Center in 2002. He became Assistant Professor in the Department of Comparative Biosciences at University of Illinois in Urbana-Champaign in 2003 and received Associate Professor tenure in 2009. Dr. Yao was recruited to National Institute of Environmental Health Sciences (NIEHS/NIH) in 2010 and was promoted to Senior Investigator in 2018. The main thrust of Dr.Yao's group is to define the normal process of how gonads and reproductive tracts form during embryogenesis and investigate the impacts of environmental exposure to harmful chemicals on these process. Dr. Yao is the corresponding author of multiple major publications on sexual differentiation of gonads and reproductive tracts, including Archambeault and Yao, PNAS, 2010 and Zhao et al., Science, 2017.
Dr. Fei Zhao is the postdoctoral fellow in Dr. Yao's lab and the first author of their recent publication in Science. Dr. Zhao received his doctoral degree at the University of Georgia in 2014 under the guidance of Dr. Xiaoqin Ye, and joined Dr. Yao's lab in 2014. Dr. Zhao received multiple awards during his postdoctoral training, including K99/R00 award in 2018.
Footnotes
Grant support: This work was supported by National Institute of Environmental Health Sciences/National Institutes of Health Intramural Research Fund Grant ZIAES102965 (to HH-CY).
References
- 1. Kobayashi A, Behringer RR. Developmental genetics of the female reproductive tract in mammals. Nat Rev Genet 2003; 4:969–980. [DOI] [PubMed] [Google Scholar]
- 2. Jost A. Recherches sur la differenciation sexuelle de lembryon de lapin .1. introduction et embryologie genitale normale. Arch Anat Microsc Morphol Exp 1947; 36:151–200. [Google Scholar]
- 3. Jost A. Problems of fetal endocrinology—the gonadal and hypophyseal hormones. Recent Prog Horm Res 1953; 8:379–418. [Google Scholar]
- 4. Gallagher TF, Koch FC. The testicular hormone. J Biol Chem 1929; 84:495–500. [Google Scholar]
- 5. Kenyon AT. The effect of testosterone propionate on the genitalia, prostate, secondary sex characters, and body weight in eunuchoidism. Endocrinology 1938; 23:121–134. [Google Scholar]
- 6. Hoberman JM, Yesalis CE. The history of synthetic testosterone. Sci Am 1995; 272:76–81. [DOI] [PubMed] [Google Scholar]
- 7. Morris JM. The syndrome of testicular feminization in male pseudohermaphrodites. Am J Obstet Gynecol 1953; 65:1192–1211. [DOI] [PubMed] [Google Scholar]
- 8. Short RV. Reproduction. Annu Rev Physiol 1967; 29:373–400. [DOI] [PubMed] [Google Scholar]
- 9. Vanha-Perttula T, Bardin CW, Allison JE, Gunbreck LG, Stanley AJ. “Testicular feminization” in the rat: morphology of the testis. Endocrinology 1970; 87:611–619. [DOI] [PubMed] [Google Scholar]
- 10. Lyon MF, Hawkes SG. X-linked gene for testicular feminization in the mouse. Nature 1970; 227:1217–1219. [DOI] [PubMed] [Google Scholar]
- 11. Chang CS, Kokontis J, Liao ST. Molecular cloning of human and rat complementary DNA encoding androgen receptors. Science 1988; 240:324–326. [DOI] [PubMed] [Google Scholar]
- 12. Lubahn DB, Joseph DR, Sullivan PM, Willard HF, French FS, Wilson EM. Cloning of human androgen receptor complementary DNA and localization to the X chromosome. Science 1988; 240:327–330. [DOI] [PubMed] [Google Scholar]
- 13. He WW, Kumar MV, Tindall DJ. A frame-shift mutation in the androgen receptor gene causes complete androgen insensitivity in the testicularfeminized mouse. Nucleic Acids Res 1991; 19:2373–2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sever R, Glass CK. Signaling by nuclear receptors. Cold Spring Harb Persp Biol 2013; 5:a016709–a016709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Murashima A, Miyagawa S, Ogino Y, Nishida-Fukuda H, Araki K, Matsumoto T, Kaneko T, Yoshinaga K, Yamamura K, Kurita T, Kato S, Moon AM et al.. Essential roles of androgen signaling in Wolffian duct stabilization and epididymal cell differentiation. Endocrinology 2011; 152:1640–1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Bentvelsen FM, Brinkmann AO, van der Schoot P, van der Linden JE, van der Kwast TH, Boersma WJ, Schroder FH, Nijman JM. Developmental pattern and regulation by androgens of androgen receptor expression in the urogenital tract of the rat. Mol Cell Endocrinol 1995; 113:245–253. [DOI] [PubMed] [Google Scholar]
- 17. Sajjad Y, Quenby SM, Nickson P, Lewis-Jones DI, Vince G. Expression of androgen receptors in upper human fetal reproductive tract. Hum Reprod 2004; 19:1659–1665. [DOI] [PubMed] [Google Scholar]
- 18. Scott HM, Mason JI, Sharpe RM. Steroidogenesis in the fetal testis and its susceptibility to disruption by exogenous compounds. Endocr Rev 2009; 30:883–925. [DOI] [PubMed] [Google Scholar]
- 19. Welsh M, Saunders PT, Sharpe RM. The critical time window for androgen-dependent development of the Wolffian duct in the rat. Endocrinology 2007; 148:3185–3195. [DOI] [PubMed] [Google Scholar]
- 20. Welsh M, Saunders PT, Marchetti NI, Sharpe RM. Androgen-dependent mechanisms of Wolffian duct development and their perturbation by flutamide. Endocrinology 2006; 147:4820–4830. [DOI] [PubMed] [Google Scholar]
- 21. Welsh M, Saunders PT, Fisken M, Scott HM, Hutchison GR, Smith LB, Sharpe RM. Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism. J Clin Invest 2008; 118:1479–1490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Gaspar ML, Meo T, Bourgarel P, Guenet JL, Tosi M. A single base deletion in the Tfm androgen receptor gene creates a short-lived messenger RNA that directs internal translation initiation. Proc Natl Acad Sci USA 1991; 88:8606–8610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yeh S, Tsai MY, Xu Q, Mu XM, Lardy H, Huang KE, Lin H, Yeh SD, Altuwaijri S, Zhou X, Xing L, Boyce BF et al.. Generation and characterization of androgen receptor knockout (ARKO) mice: An in vivo model for the study of androgen functions in selective tissues. Proc Natl Acad Sci USA 2002; 99:13498–13503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Higgins SJ, Young P, Cunha GR. Induction of functional cytodifferentiation in the epithelium of tissue recombinants. II. Instructive induction of Wolffian duct epithelia by neonatal seminal vesicle mesenchyme. Development 1989; 106:235–250. [DOI] [PubMed] [Google Scholar]
- 25. Gupta C, Singh M. Stimulation of epidermal growth factor gene expression during the fetal mouse reproductive tract differentiation: role of androgen and its receptor. Endocrinology 1996; 137:705–711. [DOI] [PubMed] [Google Scholar]
- 26. Gupta C. The role of epidermal growth factor receptor (EGFR) in male reproductive tract differentiation: stimulation of EGFR expression and inhibition of Wolffian duct differentiation with anti-EGFR antibody. Endocrinology 1996; 137:905–910. [DOI] [PubMed] [Google Scholar]
- 27. Gupta C, Siegel S, Ellis D. The role of EGF in testosterone-induced reproductive tract differentiation. Dev Biol 1991; 146:106–116. [DOI] [PubMed] [Google Scholar]
- 28. Lee D, Cross SH, Strunk KE, Morgan JE, Bailey CL, Jackson IJ, Threadgill DW. Wa5 is a novel ENU-induced antimorphic allele of the epidermal growth factor receptor. Mamm Genome 2004; 15:525–536. [DOI] [PubMed] [Google Scholar]
- 29. Luetteke NC, Qiu TH, Fenton SE, Troyer KL, Riedel RF, Chang A, Lee DC. Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. Development 1999; 126:2739–2750. [DOI] [PubMed] [Google Scholar]
- 30. Finch PW, Cunha GR, Rubin JS, Wong J, Ron D. Pattern of keratinocyte growth factor and keratinocyte growth factor receptor expression during mouse fetal development suggests a role in mediating morphogenetic mesenchymal-epithelial interactions. Dev Dyn 1995; 203:223–240. [DOI] [PubMed] [Google Scholar]
- 31. Thomson AA, Cunha GR. Prostatic growth and development are regulated by FGF10. Development 1999; 126:3693–3701. [DOI] [PubMed] [Google Scholar]
- 32. Zhao F, Franco HL, Rodriguez KF, Brown PR, Tsai MJ, Tsai SY, Yao HH. Elimination of the male reproductive tract in the female embryo is promoted by COUP-TFII in mice. Science 2017; 357:717–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Okazawa M, Murashima A, Harada M, Nakagata N, Noguchi M, Morimoto M, Kimura T, Ornitz DM, Yamada G. Region-specific regulation of cell proliferation by FGF receptor signaling during the Wolffian duct development. Dev Biol 2015; 400:139–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Nguyen AP, Chandorkar A, Gupta C. The role of growth hormone in fetal mouse reproductive tract differentiation. Endocrinology 1996; 137:3659–3666. [DOI] [PubMed] [Google Scholar]
- 35. Baker J, Hardy MP, Zhou J, Bondy C, Lupu F, Bellve AR, Efstratiadis A. Effects of an Igf1 gene null mutation on mouse reproduction. Mol Endocrinol 1996; 10:903–918. [DOI] [PubMed] [Google Scholar]
- 36. Sanford LP, Ormsby I, Gittenberger-de Groot AC, Sariola H, Friedman R, Boivin GP, Cardell EL, Doetschman T. TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 1997; 124:2659–2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Miyamoto N, Yoshida M, Kuratani S, Matsuo I, Aizawa S. Defects of urogenital development in mice lacking Emx2. Development 1997; 124:1653–1664. [DOI] [PubMed] [Google Scholar]
- 38. Torres M, Gomez-Pardo E, Dressler GR, Gruss P. Pax-2 controls multiple steps of urogenital development. Development 1995; 121:4057–4065. [DOI] [PubMed] [Google Scholar]
- 39. Cate RL, Mattaliano RJ, Hession C, Tizard R, Farber NM, Cheung A, Ninfa EG, Frey AZ, Gash DJ, Chow EP et al.. Isolation of the bovine and human genes for Mullerian inhibiting substance and expression of the human gene in animal cells. Cell 1986; 45:685–698. [DOI] [PubMed] [Google Scholar]
- 40. Josso N, Picard JY, Rey R, di Clemente N. Testicular anti-Mullerian hormone: history, genetics, regulation and clinical applications. Pediatr Endocrinol Rev 2006; 3:347–358. [PubMed] [Google Scholar]
- 41. Picard JY, Josso N. Purification of testicular anti-Mullerian hormone allowing direct visualization of the pure glycoprotein and determination of yield and purification factor. Mol Cell Endocrinol 1984; 34:23–29. [DOI] [PubMed] [Google Scholar]
- 42. Budzik GP, Donahoe PK, Hutson JM. A possible purification of Mullerian inhibiting substance and a model for its mechanism of action. Prog Clin Biol Res 1985; 171:207–223. [PubMed] [Google Scholar]
- 43. Picard JY, Benarous R, Guerrier D, Josso N, Kahn A. Cloning and expression of cDNA for anti-Mullerian hormone. Proc Natl Acad Sci USA 1986; 83:5464–5468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Behringer RR, Cate RL, Froelick GJ, Palmiter RD, Brinster RL. Abnormal sexual development in transgenic mice chronically expressing Mullerian inhibiting substance. Nature 1990; 345:167–170. [DOI] [PubMed] [Google Scholar]
- 45. Behringer RR, Finegold MJ, Cate RL. Mullerian-inhibiting substance function during mammalian sexual development. Cell 1994; 79:415–425. [DOI] [PubMed] [Google Scholar]
- 46. Picard JY, Cate RL, Racine C, Josso N. The Persistent Mullerian duct syndrome: an update based upon a personal experience of 157 cases. Sex Dev 2017; 11:109–125. [DOI] [PubMed] [Google Scholar]
- 47. Heldin CH, Moustakas A. Signaling Receptors for TGF-beta family members. Cold Spring Harb Perspect Biol 2016; 8:a022053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Baarends WM, van Helmond MJ, Post M, van der Schoot PJ, Hoogerbrugge JW, de Winter JP, Uilenbroek JT, Karels B, Wilming LG, Meijers JH et al.. A novel member of the transmembrane serine/threonine kinase receptor family is specifically expressed in the gonads and in mesenchymal cells adjacent to the Mullerian duct. Development 1994; 120:189–197. [DOI] [PubMed] [Google Scholar]
- 49. di Clemente N, Wilson C, Faure E, Boussin L, Carmillo P, Tizard R, Picard JY, Vigier B, Josso N, Cate R. Cloning, expression, and alternative splicing of the receptor for anti-Mullerian hormone. Mol Endocrinol 1994; 8:1006–1020. [DOI] [PubMed] [Google Scholar]
- 50. Teixeira J, He WW, Shah PC, Morikawa N, Lee MM, Catlin EA, Hudson PL, Wing J, Maclaughlin DT, Donahoe PK. Developmental expression of a candidate Mullerian inhibiting substance type II receptor. Endocrinology 1996; 137:160–165. [DOI] [PubMed] [Google Scholar]
- 51. Arango NA, Kobayashi A, Wang Y, Jamin SP, Lee HH, Orvis GD, Behringer RR. A mesenchymal perspective of mullerian duct differentiation and regression inAmhr2-lacZ mice. Mol Reprod Dev 2008; 75:1154–1162. [DOI] [PubMed] [Google Scholar]
- 52. Klattig J, Sierig R, Kruspe D, Besenbeck B, Englert C. Wilms' tumor protein Wt1 is an activator of the anti-Mullerian hormone receptor gene Amhr2. Mol Cell Biol 2007; 27:4355–4364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Mishina Y, Rey R, Finegold MJ, Matzuk MM, Josso N, Cate RL, Behringer RR. Genetic analysis of the Mullerian-inhibiting substance signal transduction pathway in mammalian sexual differentiation. Genes Dev 1996; 10:2577–2587. [DOI] [PubMed] [Google Scholar]
- 54. Imbeaud S, Faure E, Lamarre I, Mattei MG, di Clemente N, Tizard R, Carre-Eusebe D, Belville C, Tragethon L, Tonkin C, Nelson J, McAuliffe M et al.. Insensitivity to anti-mullerian hormone due to a mutation in the human anti-Mullerian hormone receptor. Nat Genet 1995; 11:382–388. [DOI] [PubMed] [Google Scholar]
- 55. Jamin SP, Arango NA, Mishina Y, Hanks MC, Behringer RR. Requirement of Bmpr1a for mullerian duct regression during male sexual development. Nat Genet 2002; 32:408–410. [DOI] [PubMed] [Google Scholar]
- 56. Jamin SP, Arango NA, Mishina Y, Hanks MC, Behringer RR. Genetic studies of the AMH/MIS signaling pathway for mullerian duct regression. Mol Cell Endocrinol 2003; 211:15–19. [DOI] [PubMed] [Google Scholar]
- 57. Orvis GD, Jamin SP, Kwan KM, Mishina Y, Kaartinen VM, Huang S, Roberts AB, Umans L, Huylebroeck D, Zwijsen A, Wang D, Martin JF et al.. Functional redundancy of TGF-beta family type I receptors and receptor-Smads in mediating anti-Mullerian hormone-induced Mullerian duct regression in the mouse. Biol Reprod 2008; 78:994–1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Petit FG, Deng C, Jamin SP. Partial Mullerian duct retention in Smad4 conditional mutant male mice. Int J Biol Sci 2016; 12:667–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Mullen RD, Wang Y, Liu B, Moore EL, Behringer RR. Osterix functions downstream of anti-Mullerian hormone signaling to regulate mullerian duct regression. Proc Natl Acad Sci USA 2018; 115:8382–8387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Roberts LM, Visser JA, Ingraham HA. Involvement of a matrix metalloproteinase in MIS-induced cell death during urogenital development. Development 2002; 129:1487–1496. [DOI] [PubMed] [Google Scholar]
- 61. Itoh T, Ikeda T, Gomi H, Nakao S, Suzuki T, Itohara S. Unaltered secretion of beta-amyloid precursor protein in gelatinase A (matrix metalloproteinase 2)-deficient mice. J Biol Chem 1997; 272:22389–22392. [DOI] [PubMed] [Google Scholar]
- 62. Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell 2012; 149:1192–1205. [DOI] [PubMed] [Google Scholar]
- 63. Parr BA, McMahon AP. Sexually dimorphic development of the mammalian reproductive tract requires Wnt-7a. Nature 1998; 395:707–710. [DOI] [PubMed] [Google Scholar]
- 64. Kobayashi A, Stewart CA, Wang Y, Fujioka K, Thomas NC, Jamin SP, Behringer RR. β-Catenin is essential for Mullerian duct regression during male sexual differentiation. Development 2011; 138:1967–1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Miller C, Pavlova A, Sassoon DA. Differential expression patterns of Wnt genes in the murine female reproductive tract during development and the estrous cycle. Mech Dev 1998; 76:91–99. [DOI] [PubMed] [Google Scholar]
- 66. Mericskay M, Kitajewski J, Sassoon D. Wnt5a is required for proper epithelial-mesenchymal interactions in the uterus. Development 2004; 131:2061–2072. [DOI] [PubMed] [Google Scholar]
- 67. St-Jean G, Boyer A, Zamberlam G, Godin P, Paquet M, Boerboom D. Targeted ablation of Wnt4 and Wnt5a in Mullerian duct mesenchyme impedes endometrial gland development and causes partial Mullerian agenesis. Biol Reprod 2019; 100:49–60. [DOI] [PubMed] [Google Scholar]
- 68. Park JH, Tanaka Y, Arango NA, Zhang L, Benedict LA, Roh MI, Donahoe PK, Teixeira JM. Induction of WNT inhibitory factor 1 expression by Mullerian inhibiting substance/antiMullerian hormone in the Mullerian duct mesenchyme is linked to Mullerian duct regression. Dev Biol 2014; 386:227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Bovolenta P, Esteve P, Ruiz JM, Cisneros E, Lopez-Rios J. Beyond Wnt inhibition: new functions of secreted frizzled-related proteins in development and disease. J Cell Sci 2008; 121:737–746. [DOI] [PubMed] [Google Scholar]
- 70. Warr N, Siggers P, Bogani D, Brixey R, Pastorelli L, Yates L, Dean CH, Wells S, Satoh W, Shimono A, Greenfield A. Sfrp1 and Sfrp2 are required for normal male sexual development in mice. Dev Biol 2009; 326:273–284. [DOI] [PubMed] [Google Scholar]
- 71. Cox S, Smith L, Bogani D, Cheeseman M, Siggers P, Greenfield A. Sexually dimorphic expression of secreted frizzled-related (SFRP) genes in the developing mouse Mullerian duct. Mol Reprod Dev 2006; 73:1008–1016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Deutscher E, Hung-Chang Yao H. Essential roles of mesenchyme-derived beta-catenin in mouse Mullerian duct morphogenesis. Dev Biol 2007; 307:227–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lapointe E, Boyer A, Rico C, Paquet M, Franco HL, Gossen J, DeMayo FJ, Richards JS, Boerboom D. FZD1 regulates cumulus expansion genes and is required for normal female fertility in mice. Biol Reprod 2012; 87:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Wang YK, Sporle R, Paperna T, Schughart K, Francke U. Characterization and expression pattern of the frizzled gene Fzd9, the mouse homolog of FZD9 which is deleted in Williams-Beuren syndrome. Genomics 1999; 57:235–248. [DOI] [PubMed] [Google Scholar]
- 75. Ranheim EA, Kwan HC, Reya T, Wang YK, Weissman IL, Francke U. Frizzled 9 knock-out mice have abnormal B-cell development. Blood 2005; 105:2487–2494. [DOI] [PubMed] [Google Scholar]
- 76. Nunnally AP, Parr BA. Analysis of Fz10 expression in mouse embryos. Dev Genes Evol 2004; 214:144–148. [DOI] [PubMed] [Google Scholar]
- 77. Vallon M, Yuki K, Nguyen TD, Chang J, Yuan J, Siepe D, Miao Y, Essler M, Noda M, Garcia KC, Kuo CJ. A RECK-WNT7 receptor-ligand interaction enables isoform-specific regulation of Wnt bioavailability. Cell Rep 2018; 25:339–349.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Harding SD, Armit C, Armstrong J, Brennan J, Cheng Y, Haggarty B, Houghton D, Lloyd-MacGilp S, Pi X, Roochun Y, Sharghi M, Tindal C et al.. The GUDMAP database - an online resource for genitourinary research. Development 2011; 138:2845–2853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. McMahon AP, Aronow BJ, Davidson DR, Davies JA, Gaido KW, Grimmond S, Lessard JL, Little MH, Potter SS, Wilder EL, Zhang P, GUDMAP: the genitourinary developmental molecular anatomy project. J Am Soc Nephrol 2008; 19:667–671. [DOI] [PubMed] [Google Scholar]
- 80. Kikuchi A, Yamamoto H, Kishida S. Multiplicity of the interactions of Wnt proteins and their receptors. Cell Signal 2007; 19:659–671. [DOI] [PubMed] [Google Scholar]
- 81. Mullen RD, Behringer RR. Molecular genetics of Mullerian duct formation, regression and differentiation. Sex Dev 2014; 8:281–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Tanwar PS, Zhang L, Tanaka Y, Taketo MM, Donahoe PK, Teixeira JM. Focal Mullerian duct retention in male mice with constitutively activated beta-catenin expression in the Mullerian duct mesenchyme. Proc Natl Acad Sci USA 2010; 107:16142–16147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Allard S, Adin P, Gouedard L, di Clemente N, Josso N, Orgebin-Crist MC, Picard JY, Xavier F. Molecular mechanisms of hormone-mediated Mullerian duct regression: involvement of beta-catenin. Development 2000; 127:3349–3360. [DOI] [PubMed] [Google Scholar]
- 84. Qu B, Liu BR, Du YJ, Chen J, Cheng YQ, Xu W, Wang XH. Wnt/beta-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett 2014; 7:1175–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Kruse SW, Suino-Powell K, Zhou XE, Kretschman JE, Reynolds R, Vonrhein C, Xu Y, Wang L, Tsai SY, Tsai MJ, Xu HE. Identification of COUP-TFII orphan nuclear receptor as a retinoic acid-activated receptor. PLoS Biol 2008; 6:e227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Niederreither K, Fraulob V, Garnier JM, Chambon P, Dolle P. Differential expression of retinoic acid-synthesizing (RALDH) enzymes during fetal development and organ differentiation in the mouse. Mech Dev 2002; 110:165–171. [DOI] [PubMed] [Google Scholar]
- 87. Glickman SE, Short RV, Renfree MB. Sexual differentiation in three unconventional mammals: Spotted hyenas, elephants and tammar wallabies. Horm Behav 2005; 48:403–417. [DOI] [PubMed] [Google Scholar]
- 88. Renfree MB, Chew KY, Shaw G. Hormone-independent pathways of sexual differentiation. Sex Dev 2014; 8:327–336. [DOI] [PubMed] [Google Scholar]
- 89. Ribatti D, Santoiemma M. Epithelial-mesenchymal interactions: a fundamental developmental biology mechanism. Int J Dev Biol 2014; 58:303–306. [DOI] [PubMed] [Google Scholar]
- 90. Cunha GR. Stromal induction and specification of morphogenesis and cytodifferentiation of the epithelia of the Mullerian ducts and urogenital sinus during development of the uterus and vagina in mice. J Exp Zool 1976; 196:361–369. [DOI] [PubMed] [Google Scholar]
- 91. Kurita T, Cooke PS, Cunha GR. Epithelial-stromal tissue interaction in paramesonephric (Mullerian) epithelial differentiation. Dev Biol 2001; 240:194–211. [DOI] [PubMed] [Google Scholar]
- 92. Bomgardner D, Hinton BT, Turner TT. 5′ hox genes and meis 1, a hox-DNA binding cofactor, are expressed in the adult mouse epididymis. Biol Reprod 2003; 68:644–650. [DOI] [PubMed] [Google Scholar]
- 93. Fromental-Ramain C, Warot X, Lakkaraju S, Favier B, Haack H, Birling C, Dierich A, Doll eP, Chambon P. Specific and redundant functions of the paralogous Hoxa-9 and Hoxd-9 genes in forelimb and axial skeleton patterning. Development 1996; 122:461–472. [DOI] [PubMed] [Google Scholar]
- 94. Podlasek CA, Seo RM, Clemens JQ, Ma L, Maas RL, Bushman W. Hoxa-10 deficient male mice exhibit abnormal development of the accessory sex organs. Dev Dyn 1999; 214:1–12. [DOI] [PubMed] [Google Scholar]
- 95. Benson GV, Lim H, Paria BC, Satokata I, Dey SK, Maas RL. Mechanisms of reduced fertility in Hoxa-10 mutant mice: uterine homeosis and loss of maternal Hoxa-10 expression. Development 1996; 122:2687–2696. [DOI] [PubMed] [Google Scholar]
- 96. Hsieh-Li HM, Witte DP, Weinstein M, Branford W, Li H, Small K, Potter SS. Hoxa 11 structure, extensive antisense transcription, and function in male and female fertility. Development 1995; 121:1373–1385. [DOI] [PubMed] [Google Scholar]
- 97. Davis AP, Witte DP, Hsieh-Li HM, Potter SS, Capecchi MR. Absence of radius and ulna in mice lacking hoxa-11 andhoxd-11. Nature 1995; 375:791–795. [DOI] [PubMed] [Google Scholar]
- 98. Podlasek CA, Clemens JQ, Bushman W. Hoxa-13 gene mutation results in abnormal seminal vesicle and prostate development. J Urol 1999; 161:1655–1661. [PubMed] [Google Scholar]
- 99. Warot X, Fromental-Ramain C, Fraulob V, Chambon P, Dolle P. Gene dosage-dependent effects of the Hoxa-13 and Hoxd-13 mutations on morphogenesis of the terminal parts of the digestive and urogenital tracts. Development 1997; 124:4781–4791. [DOI] [PubMed] [Google Scholar]
- 100. Snyder EM, Small CL, Bomgardner D, Xu B, Evanoff R, Griswold MD, Hinton BT. Gene expression in the efferent ducts, epididymis, and vas deferens during embryonic development of the mouse. Dev Dyn 2010; 239:2479–2491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Tomaszewski J, Joseph A, Archambeault D, Yao HH. Essential roles of inhibin beta A in mouse epididymal coiling. Proc Natl Acad Sci USA 2007; 104:11322–11327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Nie X, Arend LJ. Pkd1 is required for male reproductive tract development. Mech Dev 2013; 130:567–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Kumar M, Syed SM, Taketo MM, Tanwar PS. Epithelial Wnt/beta catenin signalling is essential for epididymal coiling. Dev Biol 2016; 412:234–249. [DOI] [PubMed] [Google Scholar]
- 104. Pihlajamaa P, Sahu B, Lyly L, Aittomaki V, Hautaniemi S, Janne OA. Tissue-specific pioneer factors associate with androgen receptor cistromes and transcription programs. EMBO J 2014; 33:312–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Sonnenberg-Riethmacher E, Walter B, Riethmacher D, Godecke S, Birchmeier C. The c-ros tyrosine kinase receptor controls regionalization and differentiation of epithelial cells in the epididymis. Genes Dev 1996; 10:1184–1193. [DOI] [PubMed] [Google Scholar]
- 106. Keilhack H, Muller M, Bohmer SA, Frank C, Weidner KM, Birchmeier W, Ligensa T, Berndt A, Kosmehl H, Gunther B, Muller T, Birchmeier C et al.. Negative regulation of ros receptor tyrosine kinase signaling. J Cell Biol 2001; 152:325–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Xu B, Yang L, Hinton BT. The Role of fibroblast growth factor receptor substrate 2 (FRS2) in the regulation of two activity levels of the components of the extracellular signal-regulated kinase (ERK) pathway in the mouse epididymis. Biol Reprod 2013; 89:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Xu B, Yang L, Lye RJ, Hinton BT. p-MAPK1/3 and DUSP6 regulate epididymal cell proliferation and survival in a region-specific manner in mice. Biol Reprod 2010; 83:807–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Xu B, Washington AM, Hinton BT. PTEN signaling through RAF1 proto-oncogene serine/threonine kinase (RAF1)/ERK in the epididymis is essential for male fertility. Proc Natl Acad Sci USA 2014; 111:18643–18648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Taylor HS, Vanden Heuvel GB, Igarashi P. A conserved Hox axis in the mouse and human female reproductive system: Late establishment and persistent adult expression of the Hoxa cluster genes. Biol Reprod 1997; 57:1338–1345. [DOI] [PubMed] [Google Scholar]
- 111. Chen X, Mu Y, Li C, Li G, Zhao H, Qin Y, Chen ZJ. Mutation screening of HOXA7 and HOXA9 genes in Chinese women with Mullerian duct abnormalities. Reprod Biomed Online 2014; 29:595–599. [DOI] [PubMed] [Google Scholar]
- 112. Gendron RL, Paradis H, Hsieh-Li HM, Lee DW, Potter SS, Markoff E. Abnormal uterine stromal and glandular function associated with maternal reproductive defects in Hoxa-11 null mice. Biol Reprod 1997; 56:1097–1105. [DOI] [PubMed] [Google Scholar]
- 113. Nakajima T, Iguchi T, Sato T. Retinoic acid signaling determines the fate of uterine stroma in the mouse Mullerian duct. Proc Natl Acad Sci USA 2016; 113:14354–14359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Mendelsohn C, Lohnes D, Decimo D, Lufkin T, LeMeur M, Chambon P, Mark M. Function of the retinoic acid receptors (RARs) during development (II). Multiple abnormalities at various stages of organogenesis in RAR double mutants. Development 1994; 120:2749–2771. [DOI] [PubMed] [Google Scholar]
- 115. Kastner P, Mark M, Ghyselinck N, Krezel W, Dupe V, Grondona JM, Chambon P. Genetic evidence that the retinoid signal is transduced by heterodimeric RXR/RAR functional units during mouse development. Development 1997; 124:313–326. [DOI] [PubMed] [Google Scholar]
- 116. Franco HL, Dai D, Lee KY, Rubel CA, Roop D, Boerboom D, Jeong JW, Lydon JP, Bagchi IC, Bagchi MK, DeMayo FJ. WNT4 is a key regulator of normal postnatal uterine development and progesterone signaling during embryo implantation and decidualization in the mouse. FASEB J 2011; 25:1176–1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Vandenberg AL, Sassoon DA. Non-canonical Wnt signaling regulates cell polarity in female reproductive tract development via van gogh-like 2. Development 2009; 136:1559–1570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Shi D, Komatsu K, Hirao M, Toyooka Y, Koyama H, Tissir F, Goffinet AM, Uemura T, Fujimori T. Celsr1 is required for the generation of polarity at multiple levels of the mouse oviduct. Development 2014; 141:4558–4568. [DOI] [PubMed] [Google Scholar]
- 119. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis 2008; 4:68–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Terakawa J, Rocchi A, Serna VA, Bottinger EP, Graff JM, Kurita T. FGFR2IIIb-MAPK activity is required for epithelial cell fate decision in the lower Mullerian duct. Mol Endocrinol 2016; 30:783–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Koster MI, Kim S, Mills AA, DeMayo FJ, Roop DR. p63 is the molecular switch for initiation of an epithelial stratification program. Genes Dev 2004; 18:126–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Kurita T, Cunha GR. Roles of p63 in differentiation of Mullerian duct epithelial cells. Ann N Y Acad Sci 2001; 948:9–12. [DOI] [PubMed] [Google Scholar]
- 123. Miller C, Sassoon DA. Wnt-7a maintains appropriate uterine patterning during the development of the mouse female reproductive tract. Development 1998; 125:3201–3211. [DOI] [PubMed] [Google Scholar]
- 124. Arango NA, Szotek PP, Manganaro TF, Oliva E, Donahoe PK, Teixeira J. Conditional deletion of beta-catenin in the mesenchyme of the developing mouse uterus results in a switch to adipogenesis in the myometrium. Dev Biol 2005; 288:276–283. [DOI] [PubMed] [Google Scholar]
- 125. Batista RL, Costa EMF, Rodrigues AS, Gomes NL, Faria JA Jr, Nishi MY, Arnhold IJP, Domenice S, Mendonca BB. Androgen insensitivity syndrome: a review. Arch Endocrinol Metab 2018; 62:227–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Mongan NP, Tadokoro-Cuccaro R, Bunch T, Hughes IA. Androgen insensitivity syndrome. Best Pract Res Clin Endocrinol Metab 2015; 29:569–580. [DOI] [PubMed] [Google Scholar]
- 127. Gottlieb B, Trifiro MA. Androgen insensitivity syndrome. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, Amemiya A (eds.), GeneReviews((R)) NCBI. Seattle, WA; 1993. [PubMed] [Google Scholar]
- 128. Gaspari L, Sampaio DR, Paris F, Audran F, Orsini M, Neto JB, Sultan C. High prevalence of micropenis in 2710 male newborns from an intensive-use pesticide area of Northeastern Brazil. Int J Androl 2012; 35:253–264. [DOI] [PubMed] [Google Scholar]
- 129. Gaspari L, Paris F, Philibert P, Audran F, Orsini M, Servant N, Maimoun L, Kalfa N, Sultan C. ‘Idiopathic’ partial androgen insensitivity syndrome in 28 newborn and infant males: impact of prenatal exposure to environmental endocrine disruptor chemicals? Eur J Endocrinol 2011; 165:579–587. [DOI] [PubMed] [Google Scholar]
- 130. Stillman RJ. In utero exposure to diethylstilbestrol: adverse effects on the reproductive tract and reproductive performance in male and female offspring. Am J Obstet Gynecol 1982; 142:905–921. [DOI] [PubMed] [Google Scholar]
- 131. Mitre AI, Castilho LN, Avarese de Figueiredo A, Arap S. Persistent Mullerian duct syndrome and prostate cancer. Urology 2002; 60:698. [DOI] [PubMed] [Google Scholar]
- 132. McCroskey Z, Koen TM, Lim DJ, Divatia MK, Shen SS, Ayala AG, Ro JY. Prostatic adenocarcinoma in the setting of persistent Mullerian duct syndrome: a case report. Hum Pathol 2018; 75:125–131. [DOI] [PubMed] [Google Scholar]
- 133. George JW, Patterson AL, Tanwar PS, Kajdacsy-Balla A, Prins GS, Teixeira JM. Specific deletion of LKB1/ Stk11 in the Mullerian duct mesenchyme drives hyperplasia of the periurethral stroma and tumorigenesis in male mice. Proc Natl Acad Sci USA 2017; 114:3445–3450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Bombard DS 2nd, Mousa SA. Mayer-Rokitansky-Kuster-Hauser syndrome: complications, diagnosis and possible treatment options: a review. Gynecol Endocrinol 2014; 30:618–623. [DOI] [PubMed] [Google Scholar]
- 135. Chen MJ, Wei SY, Yang WS, Wu TT, Li HY, Ho HN, Yang YS, Chen PL. Concurrent exome-targeted next-generation sequencing and single nucleotide polymorphism array to identify the causative genetic aberrations of isolated Mayer-Rokitansky-Kuster-Hauser syndrome. Hum Reprod 2015; 30:1732–1742. [DOI] [PubMed] [Google Scholar]
- 136. Backhouse B, Hanna C, Robevska G, van den Bergen J, Pelosi E, Simons C, Koopman P, Juniarto AZ, Grover S, Faradz S, Sinclair A, Ayers K et al.. Identification of candidate genes for mayer-rokitansky-kuster-hauser syndrome using genomic approaches. Sex Dev 2019; 13:26–34. [DOI] [PubMed] [Google Scholar]
- 137. Hannas BR, Howdeshell KL, Furr J, Gray LE Jr. In utero phthalate effects in the female rat: a model for MRKH syndrome. Toxicol Lett 2013; 223:315–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Fontana L, Gentilin B, Fedele L, Gervasini C, Miozzo M. Genetics of Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome. Clin Genet 2017; 91:233–246. [DOI] [PubMed] [Google Scholar]
- 139. Hoogendam JP, Smink M. Gartner's duct cyst. N Engl J Med 2017; 376:e27. [DOI] [PubMed] [Google Scholar]
- 140. Davidson ERW, Barber MD. A gartner duct cyst masquerading as anterior vaginal prolapse. Obstet Gynecol 2017; 130:1039–1041. [DOI] [PubMed] [Google Scholar]
- 141. Tiwari U, Relia N, Shailesh F, Kaushik C. Gartner duct cyst: CT and MRI findings. J Obstet Gynecol India 2014; 64:150–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Bats AS, Metzger U, Le Frere-Belda MA, Brisa M, Lecuru F. Malignant transformation of Gartner cyst. Int J Gynecol Cancer 2009; 19:1655–1657. [DOI] [PubMed] [Google Scholar]
- 143. Dienhart MK, Sommer RJ, Peterson RE, Hirshfield AN, Silbergeld EK. Gestational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin induces developmental defects in the rat vagina. Toxicol Sci 2000; 56:141–149. [DOI] [PubMed] [Google Scholar]
- 144. Belle M, Godefroy D, Couly G, Malone SA, Collier F, Giacobini P, Chedotal A. Tridimensional visualization and analysis of early human development. Cell 2017; 169:161–173.e12. [DOI] [PubMed] [Google Scholar]
- 145. Arora R, Fries A, Oelerich K, Marchuk K, Sabeur K, Giudice LC, Laird DJ. Insights from imaging the implanting embryo and the uterine environment in three dimensions. Development 2016; 143:4749–4754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Burton JC, Wang S, Stewart CA, Behringer RR, Larina IV. High-resolution three-dimensional in vivo imaging of mouse oviduct using optical coherence tomography. Biomed Opt Express 2015; 6:2713–2723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Wang S, Larina IV. In vivo three-dimensional tracking of sperm behaviors in the mouse oviduct. Development 2018; 145:dev157685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Wang S, Syed R, Grishina OA, Larina IV. Prolonged in vivo functional assessment of the mouse oviduct using optical coherence tomography through a dorsal imaging window. J Biophotonics 2018; 11:e201700316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Cunha GR, Robboy SJ, Kurita T, Isaacson D, Shen J, Cao M, Baskin LS. Development of the human female reproductive tract. Differentiation 2018; 103:46–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Krutsiak VN, Kumka MM. [Development of the ductule system of the epididymis in the prenatal period of human ontogeny]. Arkh Anat Gistol Embriol 1988; 95:74–78. [PubMed] [Google Scholar]
- 151. Murashima A, Xu B, Hinton BT. Understanding normal and abnormal development of the Wolffian/epididymal duct by using transgenic mice. Asian J Androl 2015; 17:749–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Rossi G, Manfrin A, Lutolf MP. Progress and potential in organoid research. Nat Rev Genet 2018; 19:671–687. [DOI] [PubMed] [Google Scholar]
- 153. Xie Y, Park ES, Xiang D, Li Z. Long-term organoid culture reveals enrichment of organoid-forming epithelial cells in the fimbrial portion of mouse fallopian tube. Stem Cell Res 2018; 32:51–60. [DOI] [PubMed] [Google Scholar]
- 154. Kessler M, Hoffmann K, Brinkmann V, Thieck O, Jackisch S, Toelle B, Berger H, Mollenkopf HJ, Mangler M, Sehouli J, Fotopoulou C, Meyer TF. The Notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nat Commun 2015; 6:8989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Turco MY, Gardner L, Hughes J, Cindrova-Davies T, Gomez MJ, Farrell L, Hollinshead M, Marsh SGE, Brosens JJ, Critchley HO, Simons BD, Hemberger M et al.. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nat Cell Biol 2017; 19:568–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Boretto M, Cox B, Noben M, Hendriks N, Fassbender A, Roose H, Amant F, Timmerman D, Tomassetti C, Vanhie A, Meuleman C, Ferrante M et al.. Development of organoids from mouse and human endometrium showing endometrial epithelium physiology and long-term expandability. Development 2017; 144:1775–1786. [DOI] [PubMed] [Google Scholar]
- 157. Yucer N, Holzapfel M, Jenkins Vogel T, Lenaeus L, Ornelas L, Laury A, Sareen D, Barrett R, Karlan BY, Svendsen CN. Directed differentiation of human induced pluripotent stem cells into fallopian tube epithelium. Sci Rep 2017; 7:10741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Miyazaki K, Dyson MT, Coon VJ, Furukawa Y, Yilmaz BD, Maruyama T, Bulun SE. Generation of progesterone-responsive endometrial stromal fibroblasts from human induced pluripotent stem cells: role of the WNT/CTNNB1 pathway. Stem Cell Rep 2018; 11:1136–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Hannema SE, Hughes IA. Regulation of Wolffian duct development. Horm Res 2007; 67:142–151. [DOI] [PubMed] [Google Scholar]
- 160. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr Protoc Mol Biol 2015; 109: 21–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Denker A, de Laat W. The second decade of 3C technologies: detailed insights into nuclear organization. Genes Dev 2016; 30:1357–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Corces MR, Trevino AE, Hamilton EG, Greenside PG, Sinnott-Armstrong NA, Vesuna S, Satpathy AT, Rubin AJ, Montine KS, Wu B, Kathiria A, Cho SW et al.. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat Methods 2017; 14:959–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Teng CS, Teng CT. Studies on sex-organ development. Changes in chemical composition and oestradiol-binding capacity in chromatin during the differentiation of chick Mullerian ducts. Biochem J 1978; 172:361–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Wu B, An C, Li Y, Yin Z, Gong L, Li Z, Liu Y, Heng BC, Zhang D, Ouyang H, Zou X. Reconstructing lineage hierarchies of mouse uterus epithelial development using single-cell analysis. Stem Cell Rep 2017; 9:381–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Mucenski ML, Mahoney R, Adam M, Potter AS, Potter SS. Single cell RNA-seq study of wild type and Hox9,10,11 mutant developing uterus. Sci Rep 2019; 9:4557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Woodworth MB, Girskis KM, Walsh CA. Building a lineage from single cells: genetic techniques for cell lineage tracking. Nat Rev Genet 2017; 18:230–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Ghosh A, Syed SM, Tanwar PS. In vivo genetic cell lineage tracing reveals that oviductal secretory cells self-renew and give rise to ciliated cells. Development 2017; 144:3031–3041. [DOI] [PubMed] [Google Scholar]
- 168. Prunskaite-Hyyrylainen R, Skovorodkin I, Xu Q, Miinalainen I, Shan J, Vainio SJ. Wnt4 coordinates directional cell migration and extension of the Mullerian duct essential for ontogenesis of the female reproductive tract. Hum Mol Genet 2016; 25:1059–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. McKenna A, Findlay GM, Gagnon JA, Horwitz MS, Schier AF, Shendure J. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science 2016; 353:aaf7907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Kalhor R, Kalhor K, Mejia L, Leeper K, Graveline A, Mali P, Church GM. Developmental barcoding of whole mouse via homing CRISPR. Science 2018; 361:eaat9804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Spanjaard B, Hu B, Mitic N, Olivares-Chauvet P, Janjuha S, Ninov N, Junker JP. Simultaneous lineage tracing and cell-type identification using CRISPR-Cas9-induced genetic scars. Nat Biotechnol 2018; 36:469–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Alemany A, Florescu M, Baron CS, Peterson-Maduro J, van Oudenaarden A. Whole-organism clone tracing using single-cell sequencing. Nature 2018; 556:108–112. [DOI] [PubMed] [Google Scholar]
- 173. Raj B, Wagner DE, McKenna A, Pandey S, Klein AM, Shendure J, Gagnon JA, Schier AF. Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain. Nat Biotechnol 2018; 36:442–450. [DOI] [PMC free article] [PubMed] [Google Scholar]



