Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
FASEB J ; 38(9): e23632, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38686936

ABSTRACT

The upper Müllerian duct (MD) is patterned and specified into two morphologically and functionally distinct organs, the oviduct and uterus. It is known that this regionalization process is instructed by inductive signals from the adjacent mesenchyme. However, the interaction landscape between epithelium and mesenchyme during upper MD development remains largely unknown. Here, we performed single-cell transcriptomic profiling of mouse neonatal oviducts and uteri at the initiation of MD epithelial differentiation (postnatal day 3). We identified major cell types including epithelium, mesenchyme, pericytes, mesothelium, endothelium, and immune cells in both organs with established markers. Moreover, we uncovered region-specific epithelial and mesenchymal subpopulations and then deduced region-specific ligand-receptor pairs mediating mesenchymal-epithelial interactions along the craniocaudal axis. Unexpectedly, we discovered a mesenchymal subpopulation marked by neurofilaments with specific localizations at the mesometrial pole of both the neonatal oviduct and uterus. Lastly, we analyzed and revealed organ-specific signature genes of pericytes and mesothelial cells. Taken together, our study enriches our knowledge of upper MD development, and provides a manageable list of potential genes, pathways, and region-specific cell subtypes for future functional studies.


Subject(s)
Mullerian Ducts , Oviducts , Single-Cell Analysis , Transcriptome , Uterus , Animals , Female , Mice , Uterus/metabolism , Uterus/cytology , Mullerian Ducts/metabolism , Oviducts/metabolism , Oviducts/cytology , Gene Expression Profiling , Animals, Newborn , Cell Differentiation , Mesoderm/metabolism , Mesoderm/cytology , Epithelial Cells/metabolism , Mice, Inbred C57BL , Gene Expression Regulation, Developmental
2.
J Clin Invest ; 133(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37847567

ABSTRACT

Three sisters, born from consanguineous parents, manifested a unique Müllerian anomaly characterized by uterine hypoplasia with thin estrogen-unresponsive endometrium and primary amenorrhea, but with spontaneous tubal pregnancies. Through whole-exome sequencing followed by comprehensive genetic analysis, a missense variant was identified in the OSR1 gene. We therefore investigated OSR1/OSR1 expression in postpubertal human uteri, and the prenatal and postnatal expression pattern of Osr1/Osr1 in murine developing Müllerian ducts (MDs) and endometrium, respectively. We then investigated whether Osr1 deletion would affect MD development, using WT and genetically engineered mice. Human uterine OSR1/OSR1 expression was found primarily in the endometrium. Mouse Osr1 was expressed prenatally in MDs and Wolffian ducts (WDs), from rostral to caudal segments, in E13.5 embryos. MDs and WDs were absent on the left side and MDs were rostrally truncated on the right side of E13.5 Osr1-/- embryos. Postnatally, Osr1 was expressed in mouse uteri throughout their lifespan, peaking at postnatal days 14 and 28. Osr1 protein was present primarily in uterine luminal and glandular epithelial cells and in the epithelial cells of mouse oviducts. Through this translational approach, we demonstrated that OSR1 in humans and mice is important for MD development and endometrial receptivity and may be implicated in uterine factor infertility.


Subject(s)
Infertility , Mullerian Ducts , Animals , Female , Humans , Mice , Pregnancy , Endometrium , Epithelial Cells , Mullerian Ducts/metabolism , Uterus
3.
Front Endocrinol (Lausanne) ; 13: 916173, 2022.
Article in English | MEDLINE | ID: mdl-35909540

ABSTRACT

Dlx5 and Dlx6 encode distal-less homeodomain transcription factors that are present in the genome as a linked pair at a single locus. Dlx5 and Dlx6 have redundant roles in craniofacial, skeletal, and uterine development. Previously, we performed a transcriptome comparison for anti-Müllerian hormone (AMH)-induced genes expressed in the Müllerian duct mesenchyme of male and female mouse embryos. In that study, we found that Dlx5 transcripts were nearly seven-fold higher in males compared to females and Dlx6 transcripts were found only in males, suggesting they may be AMH-induced genes. Therefore, we investigated the role of Dlx5 and Dlx6 during AMH-induced Müllerian duct regression. We found that Dlx5 was detected in the male Müllerian duct mesenchyme from E14.5 to E16.5. In contrast, in female embryos Dlx5 was detected in the Müllerian duct epithelium. Dlx6 expression in Müllerian duct mesenchyme was restricted to males. Dlx6 expression was not detected in female Müllerian duct mesenchyme or epithelium. Genetic experiments showed that AMH signaling is necessary for Dlx5 and Dlx6 expression. Müllerian duct regression was variable in Dlx5 homozygous mutant males at E16.5, ranging from regression like controls to a block in Müllerian duct regression. In E16.5 Dlx6 homozygous mutants, Müllerian duct tissue persisted primarily in the region adjacent to the testes. In Dlx5-6 double homozygous mutant males Müllerian duct regression was also found to be incomplete but more severe than either single mutant. These studies suggest that Dlx5 and Dlx6 act redundantly to mediate AMH-induced Müllerian duct regression during male differentiation.


Subject(s)
Genes, Homeobox , Mullerian Ducts , Animals , DNA-Binding Proteins/genetics , Female , Homeodomain Proteins/genetics , Male , Mice , Mullerian Ducts/metabolism , Sex Differentiation , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Front Endocrinol (Lausanne) ; 13: 905324, 2022.
Article in English | MEDLINE | ID: mdl-35721723

ABSTRACT

Over seventy years ago it was proposed that the fetal testis produces a hormone distinct from testosterone that is required for complete male sexual development. At the time the hormone had not yet been identified but was invoked by Alfred Jost to explain why the Müllerian duct, which develops into the female reproductive tract, regresses in the male fetus. That hormone, anti-Müllerian hormone (AMH), and its specific receptor, AMHR2, have now been extensively characterized and belong to the transforming growth factor-ß families of protein ligands and receptors involved in growth and differentiation. Much is now known about the downstream events set in motion after AMH engages AMHR2 at the surface of specific Müllerian duct cells and initiates a cascade of molecular interactions that ultimately terminate in the nucleus as activated transcription factors. The signals generated by the AMH signaling pathway are then integrated with signals coming from other pathways and culminate in a complex gene regulatory program that redirects cellular functions and fates and leads to Müllerian duct regression.


Subject(s)
Anti-Mullerian Hormone , Peptide Hormones , Anti-Mullerian Hormone/metabolism , Female , Humans , Male , Mullerian Ducts/metabolism , Peptide Hormones/metabolism , Protein Serine-Threonine Kinases , Sex Differentiation/genetics , Signal Transduction/physiology
5.
J Int Med Res ; 50(5): 3000605221096275, 2022 May.
Article in English | MEDLINE | ID: mdl-35574814

ABSTRACT

Mullerian cysts are benign tumors that are very rare in the posterior mediastinum. It is necessary to distinguish Mullerian cysts from benign tumors or other types of cyst in the posterior mediastinum. A 42-year-old woman visited our hospital for a routine check-up, and a mediastinal mass was identified on chest computed tomography (CT). Contrast-enhanced chest magnetic resonance imaging (MRI) revealed a 4.0 × 2.6 × 2.8-cm mass, and a neurogenic tumor or esophageal cyst was suspected. Single-port thoracoscopic surgery was performed for cyst removal. Histopathological examination of the resected tissue revealed that the cyst wall was covered with a single layer of ciliated columnar epithelium. Immunohistochemical staining revealed positivity for paired box gene 8 (PAX8), Wilms tumor protein 1 (WT-1), estrogen receptor (ER), and progesterone receptor (PR). Therefore, a diagnosis of mediastinal Mullerian cyst was made. Mediastinal Mullerian cysts should be included in the differential diagnosis of posterior mediastinal cysts. Cystic lesions in the posterior mediastinum should be removed surgically and undergo immunohistochemical examination.


Subject(s)
Mediastinal Cyst , Adult , Female , Humans , Magnetic Resonance Imaging , Mediastinal Cyst/diagnostic imaging , Mediastinal Cyst/surgery , Mediastinum/pathology , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Mullerian Ducts/surgery , Tomography, X-Ray Computed
6.
Morphologie ; 106(355): 252-259, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34924282

ABSTRACT

The anti-Müllerian hormone (AMH) is a heterodimeric glycoprotein belonging to the TGFb superfamily implicated in human embryonic development. This hormone was first described as allowing regression of the epithelial embryonic Müllerian structures in males, which would otherwise differentiate into the uterus and fallopian tubes. It activates a signaling pathway mediated by two transmembrane receptors. Binding of AMH to its receptor induces morphological changes leading to the degeneration of Müllerian ducts. Recently, new data has shown the role played by this hormone on structures other than the genital tract. If testicular AMH expression decreases in humans over the course of a lifetime, synthesis may persist in other tissues in adulthood. The mechanisms underlying its production have been unveiled. The aim of this review is to describe the different pathways in which AMH has been identified and plays a pivotal role.


Subject(s)
Anti-Mullerian Hormone , Mullerian Ducts , Male , Female , Humans , Adult , Anti-Mullerian Hormone/metabolism , Mullerian Ducts/metabolism , Glycoproteins/metabolism , Testis/metabolism , Signal Transduction/physiology
7.
J Pediatr Endocrinol Metab ; 34(10): 1247-1255, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34265880

ABSTRACT

OBJECTIVES: Antimullerian hormone (AMH) causes regression of the mullerian ducts in the male fetus. The appendix testis (AT) is a vestigial remnant of mullerian duct origin, containing both androgen (AR) and estrogen (ER) receptors. The role of both AMH and AT in testicular descent is yet to be studied. We investigated the possible association of AMH with AT size, the AR and ER, and their expression in the AT, in congenital cryptorchidism. METHODS: A total of 26 patients with congenital unilateral cryptorchidism and 26 controls with orthotopic testes were investigated, and 21 ATs were identified in each group. AMH and insulin-like three hormone (INSL3) concentrations were measured with spectrophotometry. AR and ER receptor expression was assessed with immunohistochemistry using monoclonal antibodies R441 for AR and MAB463 for ER. For the estimation of receptor expression, the Allred Score method was used. RESULTS: AMH concentrations did not present significant differences between patients with congenital cryptorchidism and the controls. Also, no correlation was found between AMH, INSL3, and AT length. Allred scores did not present significant differences. However, expression percentiles and intensity for both receptors presented significant differences. Three children with cryptorchidism and the highest AMH levels also had the highest estrogen receptor scores in the AT. CONCLUSIONS: No association was found between AMH and the studied major parameters. However, higher AMH concentrations, in combination with higher estrogen receptor scores in the AT, may play a role in cryptorchidism in some children. Larger population samples are needed to verify this observation.


Subject(s)
Anti-Mullerian Hormone/blood , Cryptorchidism/pathology , Genitalia, Male/pathology , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Child, Preschool , Cohort Studies , Cryptorchidism/blood , Cryptorchidism/genetics , Gene Expression , Genitalia, Male/abnormalities , Genitalia, Male/embryology , Greece , Humans , Infant , Insulin/blood , Male , Mullerian Ducts/abnormalities , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Organ Size , Proteins , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Testis/abnormalities , Testis/pathology
8.
Differentiation ; 118: 34-40, 2021.
Article in English | MEDLINE | ID: mdl-33707128

ABSTRACT

The development of the female reproductive tract can be divided into three parts consisting of Müllerian duct organogenesis, pre-sexual maturation organ development, and post-sexual maturation hormonal regulation. In primates, Müllerian duct organogenesis proceeds in an estrogen independent fashion based on transcriptional pathways that are suppressed in males by the presence of AMH and SRY. However, clinical experience indicates that exposure to xenoestrogens such as diethylstilbestrol (DES) during critical periods including late organogenesis and pre-sexual maturational development can have substantial effects on uterine morphology, and confer increased risk of disease states later in life. Recent evidence has demonstrated that these effects are in part due to epigenetic regulation of gene expression, both in the form of aberrant CpG methylation, and accompanying histone modifications. While xenoestrogens and selective estrogen receptor modulators (SERMS) both can induce non-canonical binding confirmations in estrogen receptors, the primate specific fetal estrogens Estriol and Estetrol may act in a similar fashion to alter gene expression through tissue specific epigenetic modulation.


Subject(s)
DNA Methylation/genetics , Estrogens/genetics , Genitalia, Female/growth & development , Organogenesis/genetics , Animals , DNA Methylation/drug effects , Diethylstilbestrol/pharmacology , Epigenesis, Genetic/genetics , Estradiol/metabolism , Estrogens/metabolism , Female , Genitalia, Female/metabolism , Humans , Mullerian Ducts/drug effects , Mullerian Ducts/growth & development , Mullerian Ducts/metabolism , Organogenesis/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Uterus/drug effects , Uterus/growth & development
9.
Int J Mol Sci ; 22(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530552

ABSTRACT

The reproductive tract in mammals emerges from two ductal systems during embryogenesis: Wolffian ducts (WDs) and Mullerian ducts (MDs). Most of the female reproductive tract (FRT) including the oviducts, uterine horn and cervix, originate from MDs. It is widely accepted that the formation of MDs depends on the preformed WDs within the urogenital primordia. Here, we found that the WD mesenchyme under the regulation of Hedgehog (Hh) signaling is closely related to the developmental processes of the FRT during embryonic and postnatal periods. Deficiency of Sonic hedgehog (Shh), the only Hh ligand expressed exclusively in WDs, prevents the MD mesenchyme from affecting uterine growth along the radial axis. The in vivo cell tracking approach revealed that after WD regression, distinct cells responding to WD-derived Hh signal continue to exist in the developing FRT and gradually contribute to the formation of various tissues such as smooth muscle, endometrial stroma and vascular vessel, in the mouse uterus. Our study thus provides a novel developmental mechanism of FRT relying on WD.


Subject(s)
Genitalia, Female/embryology , Genitalia, Female/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Signal Transduction , Uterus/embryology , Uterus/metabolism , Animals , Biomarkers , Cell Differentiation , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Knockout , Models, Biological , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Organogenesis/genetics
10.
Hum Pathol ; 109: 21-30, 2021 03.
Article in English | MEDLINE | ID: mdl-33275953

ABSTRACT

Mucinous cystadenoma is one of the most common benign ovarian neoplasms. The immunophenotypes and histogenetic relationships of mucinous cystadenomas with a Müllerian-type epithelium have not been fully explored. We elucidated the direction of differentiation of the mucinous epithelium that constitutes mucinous cystadenomas. Special attention was paid to the existence of gastrointestinal (GI)-type mucinous epithelium, and its association with background Müllerian-type epithelium. Immunohistochemistry was performed in 139 cases of mucinous cystadenoma to evaluate the expression of Claudin-18 (CLDN18), a novel marker of gastric differentiation; CDX2, a marker of intestinal differentiation; and estrogen receptor (ER), a marker of Müllerian differentiation. We found that GI differentiation characterized by CLDN18 and/or CDX2 positivity was observed in mucinous epithelium of most mucinous cystadenomas (129/139 cases, 93%). In a subset of these cases, the tumor was composed of mucinous epithelium exhibiting an intermediate GI and Müllerian phenotype (CLDN18+/CDX2±/ER+). Of note, in 12 cases, a transition from background Müllerian-type epithelium to mucinous epithelium with GI differentiation was identified. A minor subset (6%) of mucinous cystadenomas was considered a pure Müllerian type because the epithelium exhibited a CLDN18-/CDX2-/ER + immunophenotype. In conclusion, mucinous cystadenomas consist of three major subtypes: GI, Müllerian, and intermediate types. Most mucinous cystadenomas are GI-type, and they should be considered a precursor of GI-type mucinous borderline tumors. The existence of intermediate-type mucinous cystadenomas, and areas of transition from Müllerian-type to GI-type epithelium suggest that GI-type mucinous epithelium can arise from Müllerian duct derivatives or surface epithelium exhibiting Müllerian metaplasia in the ovary.


Subject(s)
Cystadenoma, Mucinous/pathology , Gastrointestinal Neoplasms/pathology , Immunohistochemistry , Ovarian Neoplasms/pathology , Adenocarcinoma, Mucinous/pathology , Adult , Biomarkers, Tumor/analysis , Cystadenoma, Mucinous/genetics , Female , Humans , Immunohistochemistry/methods , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/metabolism , Phenotype , Receptors, Estrogen/analysis
11.
Differentiation ; 118: 4-23, 2021.
Article in English | MEDLINE | ID: mdl-33189416

ABSTRACT

In the early 1960's, at Professor Bern's laboratory, University of California, Berkeley) in the US, Takasugi discovered ovary-independent, persistent vaginal changes in mice exposed neonatally to estrogen, which resulted in vaginal cancer later in life. Reproductive abnormalities in rodents were reported as a result of perinatal exposure to various estrogenic chemicals. Ten years later, vaginal cancers were reported in young women exposed in utero to the synthetic estrogen diethylstilbestrol (DES) and this has been called the "DES syndrome". The developing organism is particularly sensitive to developmental exposure to estrogens inducing long-term changes in various organs including the reproductive organs. The molecular mechanism underlying the persistent vaginal changes induced by perinatal estrogen exposure was partly demonstrated. Persistent phosphorylation and sustained expression of EGF-like growth factors, lead to estrogen receptor α (ESR1) activation, and then persistent vaginal epithelial cell proliferation. Agents which are weakly estrogenic by postnatal criteria may have major developmental effects, especially during a critical perinatal period. The present review outlines various studies conducted by four generations of investigators all under the influence of Prof. Bern. The studies include reports of persistent changes induced by neonatal androgen exposure, analyses of estrogen responsive genes, factors determining epithelial differentiation in the Müllerian duct, ESR and growth factor signaling, and polyovular follicles in mammals. This review is then expanded to the studies on the effects of environmental estrogens on wildlife and endocrine disruption in Daphnids.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogens/toxicity , Gonadal Steroid Hormones/metabolism , Vaginal Neoplasms/genetics , Animals , Animals, Newborn , Cell Proliferation/drug effects , Diethylstilbestrol/pharmacology , Estrogens/analogs & derivatives , Female , Gonadal Steroid Hormones/biosynthesis , Humans , Mice , Mullerian Ducts/drug effects , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Pregnancy , Vagina/drug effects , Vagina/metabolism , Vagina/pathology , Vaginal Neoplasms/chemically induced , Vaginal Neoplasms/pathology
12.
BMC Genomics ; 21(1): 688, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33008304

ABSTRACT

BACKGROUND: Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract in vertebrates. Many disorders of female reproduction can be attributed to anomalies of Müllerian duct development. However, the molecular genetics of Müllerian duct formation is poorly understood and most disorders of duct development have unknown etiology. In this study, we describe for the first time the transcriptional landscape of the embryonic Müllerian duct, using the chicken embryo as a model system. RNA sequencing was conducted at 1 day intervals during duct formation to identify developmentally-regulated genes, validated by in situ hybridization. RESULTS: This analysis detected hundreds of genes specifically up-regulated during duct morphogenesis. Gene ontology and pathway analysis revealed enrichment for developmental pathways associated with cell adhesion, cell migration and proliferation, ERK and WNT signaling, and, interestingly, axonal guidance. The latter included factors linked to neuronal cell migration or axonal outgrowth, such as Ephrin B2, netrin receptor, SLIT1 and class A semaphorins. A number of transcriptional modules were identified that centred around key hub genes specifying matrix-associated signaling factors; SPOCK1, HTRA3 and ADGRD1. Several novel regulators of the WNT and TFG-ß signaling pathway were identified in Müllerian ducts, including APCDD1 and DKK1, BMP3 and TGFBI. A number of novel transcription factors were also identified, including OSR1, FOXE1, PRICKLE1, TSHZ3 and SMARCA2. In addition, over 100 long non-coding RNAs (lncRNAs) were expressed during duct formation. CONCLUSIONS: This study provides a rich resource of new candidate genes for Müllerian duct development and its disorders. It also sheds light on the molecular pathways engaged during tubulogenesis, a fundamental process in embryonic development.


Subject(s)
Gene Expression Regulation, Developmental , Mullerian Ducts/metabolism , Transcriptome , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chick Embryo , Female , Mullerian Ducts/embryology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Fertil Steril ; 114(6): 1297-1305, 2020 12.
Article in English | MEDLINE | ID: mdl-33036791

ABSTRACT

OBJECTIVE: To study whether markers of prenatal exposure to reproductive hormones are related to Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome, polycystic ovary syndrome (PCOS), and endometriosis. DESIGN: Case-control study. Comparison of sex hormone-related external genital and digital characteristics in cases and controls. SETTING: University hospital. PATIENT(S): We enrolled 172 women in four groups-women with MKRH, women with PCOS, women with endometriosis, and controls (43 in each group). INTERVENTION(S): Measurement of two anthropometric biomarkers: anogenital distance and digit ratio. MAIN OUTCOME MEASURE(S): Anogenital distance was measured from the anus to the anterior clitoral surface (AGDac) and from the anus to the posterior fourchette (AGDaf). For the digit ratio we used a direct, as well as a computer-assisted graphic measurement to measure the length of the second and fourth digit. RESULT(S): After adjustment for body mass index and age, AGDac was the shortest in endometriosis and the longest in PCOS groups, with a mean difference of 10 mm (95% confidence interval 3.1-16.8). AGDaf but not AGDac measures were found to be significantly larger in the MRKH group, with a mean difference compared with controls of 2.6 mm (95% confidence interval 0.1-5.2). The digit ratio was not significantly different between the groups. CONCLUSION(S): In this study we did find limited evidence for androgen exposure during the development of MRKH. This is compatible with the hypothesis that the uterovaginal agenesis may have been the result of temporary prenatal exposure to altered gonadal hormone concentrations. For endometriosis and PCOS we confirm previously observed associations for anogenital distance reflecting possible estrogen-based and androgen-based intrauterine origins, respectively. DUTCH TRIAL REGISTRATION NUMBER: NTR7492.


Subject(s)
46, XX Disorders of Sex Development/metabolism , Anthropometry , Congenital Abnormalities/metabolism , Endometriosis/metabolism , Gonadal Hormones/metabolism , Mullerian Ducts/abnormalities , Polycystic Ovary Syndrome/metabolism , 46, XX Disorders of Sex Development/diagnosis , 46, XX Disorders of Sex Development/pathology , Adult , Anal Canal/pathology , Case-Control Studies , Clitoris/pathology , Congenital Abnormalities/diagnosis , Congenital Abnormalities/pathology , Endometriosis/diagnosis , Endometriosis/pathology , Female , Fingers/pathology , Humans , Middle Aged , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/pathology , Predictive Value of Tests , Reproducibility of Results
14.
J Endocrinol ; 244(2): 395-413, 2020 02.
Article in English | MEDLINE | ID: mdl-31829965

ABSTRACT

The embryonic Müllerian ducts give rise to the female reproductive tract (fallopian tubes, uterus and upper vagina in humans, the oviducts in birds). Embryonic Müllerian ducts initially develop in both sexes, but later regress in males under the influence of anti-Müllerian hormone. While the molecular and endocrine control of duct regression in males have been well studied, early development of the ducts in both sexes is less well understood. Here, we describe a novel role for the adhesion G protein-coupled receptor, GPR56, in development of the Müllerian ducts in the chicken embryo. GPR56 is expressed in the ducts of both sexes from early stages. The mRNA is present during the elongation phase of duct formation, and it is restricted to the inner Müllerian duct epithelium. The putative ligand, Collagen III, is abundantly expressed in the Müllerian duct at the same developmental stages. Knockdown of GPR56 expression using in ovo electroporation results in variably truncated ducts, with a loss of expression of both epithelial and mesenchymal markers of duct development. Over-expression of GPR56 in vitro results in enhanced cell proliferation and cell migration. These results show that GPR56 plays an essential role in avian Müllerian duct development through the regulation of duct elongation.


Subject(s)
Avian Proteins/metabolism , Chickens/metabolism , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Avian Proteins/genetics , Cell Proliferation , Chick Embryo , Chickens/genetics , Chickens/growth & development , Female , Gene Expression Regulation, Developmental , Male , Receptors, G-Protein-Coupled/genetics
15.
Development ; 146(20)2019 10 18.
Article in English | MEDLINE | ID: mdl-31575647

ABSTRACT

WNT signaling plays essential roles in the development and function of the female reproductive tract. Although crosstalk with the Hippo pathway is a key regulator of WNT signaling, whether Hippo itself plays a role in female reproductive biology remains largely unknown. Here, we show that conditional deletion of the key Hippo kinases Lats1 and Lats2 in mouse Müllerian duct mesenchyme cells caused them to adopt the myofibroblast cell fate, resulting in profound reproductive tract developmental defects and sterility. Myofibroblast differentiation was attributed to increased YAP and TAZ expression (but not to altered WNT signaling), leading to the direct transcriptional upregulation of Ctgf and the activation of the myofibroblast genetic program. Müllerian duct mesenchyme cells also became myofibroblasts in male mutant embryos, which impeded the development of the male reproductive tract and resulted in cryptorchidism. The inactivation of Lats1/2 in differentiated uterine stromal cells in vitro did not compromise their ability to decidualize, suggesting that Hippo is dispensable during implantation. We conclude that Hippo signaling is required to suppress the myofibroblast genetic program and maintain multipotency in Müllerian mesenchyme cells.


Subject(s)
Mullerian Ducts/cytology , Mullerian Ducts/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Chromatin Immunoprecipitation , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endometrium/cytology , Endometrium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics
16.
Biol Reprod ; 100(1): 49-60, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30010727

ABSTRACT

Wnt4 and Wnt5a have well-established roles in the embryonic development of the female reproductive tract, as well as in implantation, decidualization, and ovarian function in adult mice. Although these roles appear to overlap, whether Wnt5a and Wnt4 are functionally redundant in these tissues has not been determined. We addressed this by concomitantly inactivating Wnt4 and Wnt5a in the Müllerian mesenchyme and in ovarian granulosa cells by crossing mice bearing floxed alleles to the Amhr2cre strain. Whereas fertility was reduced by ∼50% in Wnt4flox/flox; Amhr2cre/+ and Wnt5aflox/flox; Amhr2cre/+ females, Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice were either nearly or completely sterile. Loss of fertility was not due to an ovarian defect, as serum ovarian hormone levels, follicle counts, and ovulation rates were comparable to controls. Conversely, the uterus was abnormal in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice, with thin myometrial and stromal layers, frequent fibrosis and a >90% reduction in numbers of uterine glands, suggesting redundant or additive roles of Wnt4 and Wnt5a in uterine adenogenesis. Loss of fertility in Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ mice was attributed to defects in decidualization, implantation, and placental development, the severity of which were proportional to the extent of gland loss. Furthermore, a third of Wnt4flox/flox; Wnt5aflox/flox; Amhr2cre/+ females had a partial agenesis of Müllerian duct-derived structures, but with normal oviducts and ovaries. Together, our results suggest that Wnt4 and Wnt5a play redundant roles in the development of the female reproductive tract, and may provide insight into the etiology of certain cases of Müllerian agenesis in women.


Subject(s)
Mullerian Ducts/abnormalities , Mullerian Ducts/metabolism , Urogenital Abnormalities/genetics , Uterus/abnormalities , Wnt-5a Protein/genetics , Wnt4 Protein/genetics , Animals , Female , Gene Deletion , Infertility, Female/embryology , Infertility, Female/genetics , Infertility, Female/metabolism , Male , Mesoderm/abnormalities , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Transgenic , Mullerian Ducts/pathology , Myometrium/abnormalities , Myometrium/metabolism , Pregnancy , Urogenital Abnormalities/metabolism , Urogenital Abnormalities/pathology , Uterus/metabolism , Wnt-5a Protein/metabolism , Wnt4 Protein/metabolism
17.
Genetics ; 211(1): 219-233, 2019 01.
Article in English | MEDLINE | ID: mdl-30446521

ABSTRACT

In laboratory strains of zebrafish, sex determination occurs in the absence of a typical sex chromosome and it is not known what regulates the proportion of animals that develop as males or females. Many sex determination and gonad differentiation genes that act downstream of a sex chromosome are well conserved among vertebrates, but studies that test their contribution to this process have mostly been limited to mammalian models. In mammals, WNT4 is a signaling ligand that is essential for ovary and Müllerian duct development, where it antagonizes the male-promoting FGF9 signal. Wnt4 is well conserved across all vertebrates, but it is not known if Wnt4 plays a role in sex determination and/or the differentiation of sex organs in nonmammalian vertebrates. This question is especially interesting in teleosts, such as zebrafish, because they lack an Fgf9 ortholog. Here we show that wnt4a is the ortholog of mammalian Wnt4, and that wnt4b was present in the last common ancestor of humans and zebrafish, but was lost in mammals. We show that wnt4a loss-of-function mutants develop predominantly as males and conclude that wnt4a activity promotes female sex determination and/or differentiation in zebrafish. Additionally, both male and female wnt4a mutants are sterile due to defects in reproductive duct development. Together these results strongly argue that Wnt4a is a conserved regulator of female sex determination and reproductive duct development in mammalian and nonmammalian vertebrates.


Subject(s)
Mullerian Ducts/metabolism , Sex Differentiation , Wnt4 Protein/genetics , Zebrafish Proteins/genetics , Animals , Female , Fibroblast Growth Factor 9/genetics , Fibroblast Growth Factor 9/metabolism , Male , Mullerian Ducts/embryology , Wnt4 Protein/metabolism , Zebrafish , Zebrafish Proteins/metabolism
18.
Am J Surg Pathol ; 42(12): 1596-1606, 2018 12.
Article in English | MEDLINE | ID: mdl-30148742

ABSTRACT

Mesonephric carcinomas of the gynecologic tract are neoplasms that are often under-recognized due to their varied morphologic appearances. Recently, GATA3 and TTF1 have been reported to be useful immunohistochemical markers for distinguishing mesonephric carcinomas from its morphologic mimics. Herein, we compared the performance of GATA3 and TTF1 to the traditional markers used for mesonephric carcinomas, CD10 and calretinin. We studied 694 cases: 8 mesonephric carcinomas (7 cervical [includes 3 mesonephric carcinosarcomas], 1 vaginal), 5 mesonephric-like carcinomas (4 uterine corpus, 1 ovarian), 585 endometrial adenocarcinomas, and 96 cervical adenocarcinomas. Mesonephric-like carcinomas were defined as tumors exhibiting the classic morphologic features of mesonephric carcinoma, but occurring outside of the cervix and without convincing mesonephric remnants. GATA3 had the highest sensitivity and specificity (91% and 94%) compared with TTF1 (45% and 99%), CD10 (73% and 83%), and calretinin (36% and 89%). GATA3, however, also stained a substantial number of uterine carcinosarcomas (23/113, 20%). TTF1 was positive in 5/5 (100%) mesonephric-like carcinomas and only 1/8 (13%) mesonephric carcinomas. In 4/6 (67%) TTF1 positive cases, GATA3 exhibited an inverse staining pattern with TTF1. In summary, GATA3 was the best overall marker for mesonephric and mesonephric-like carcinomas, but cannot be used to distinguish mesonephric carcinosarcomas from Müllerian carcinosarcomas. The inverse staining pattern between GATA3 and TTF1, suggests that TTF1 may be useful when GATA3 is negative in small biopsies where mesonephric or mesonephric-like carcinoma is suspected. The greater TTF1 positivity in mesonephric-like carcinomas suggests they may be biologically different from prototypical mesonephric carcinomas.


Subject(s)
Adenocarcinoma/chemistry , Biomarkers, Tumor/analysis , Calbindin 2/analysis , Carcinosarcoma/chemistry , Endometrial Neoplasms/chemistry , GATA3 Transcription Factor/analysis , Mullerian Ducts/chemistry , Neprilysin/analysis , Thyroid Nuclear Factor 1/analysis , Uterine Cervical Neoplasms/chemistry , Vaginal Neoplasms/chemistry , Wolffian Ducts/chemistry , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Biopsy , Carcinosarcoma/pathology , Diagnosis, Differential , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Mullerian Ducts/metabolism , Predictive Value of Tests , Tissue Array Analysis , Uterine Cervical Neoplasms/pathology , Vaginal Neoplasms/pathology , Wolffian Ducts/pathology
19.
Fertil Steril ; 109(3): 371-379, 2018 03.
Article in English | MEDLINE | ID: mdl-29566849

ABSTRACT

Adenomyosis is a commonly diagnosed estrogen-dependent gynecological disorder that causes pelvic pain, abnormal uterine bleeding, and infertility. Despite its prevalence and severity of symptoms, its pathogenesis and etiology have not yet been elucidated. The aim of this manuscript is to review the different hypotheses on the origin of adenomyotic lesions and the mechanisms involved in the evolution and progression of the disease. Two main theories have been proposed to explain the origin of adenomyosis. The most common suggests involvement of tissue injury and the repair mechanism and claims that adenomyosis results from invagination of the endometrial basalis into the myometrium. An alternative theory maintains that adenomyotic lesions result from metaplasia of displaced embryonic pluripotent Müllerian remnants or differentiation of adult stem cells. Previous investigations performed in human adenomyotic lesions and corroborated by studies in mice supported the involvement of the epithelial-mesenchymal transition process in the early stages of progression and spread of adenomyosis. However, studies conducted in a recently developed baboon model indicate that collective cell migration may be implicated in the later events of invasion. This suggests that the invasiveness of this complex uterine disorder is not driven by a single mechanism of migration but by a time-dependent combination of two processes.


Subject(s)
Adenomyosis/pathology , Cell Movement , Uterine Diseases/pathology , Uterus/pathology , Adenomyosis/genetics , Adenomyosis/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Disease Progression , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Estradiol/metabolism , Female , Humans , Metaplasia , Mullerian Ducts/metabolism , Mullerian Ducts/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Signal Transduction , Time Factors , Uterine Diseases/genetics , Uterine Diseases/metabolism , Uterus/metabolism
20.
Wiley Interdiscip Rev Dev Biol ; 7(3): e310, 2018 05.
Article in English | MEDLINE | ID: mdl-29350886

ABSTRACT

The Müllerian ducts are part of the embryonic urogenital system. They give rise to mature structures that serve a critical function in the transport and development of the oocyte and/or embryo. In most vertebrates, both sexes initially develop Müllerian ducts during embryogenesis, but they regress in males under the influence of testis-derived Anti-Müllerian Hormone (AMH). A number of regulatory factors have been shown to be essential for proper duct development, including Bmp and Wnt signaling molecules, together with homeodomain transcription factors such as PAX2 and LIM1. Later in development, the fate of the ducts diverges between males and females and is regulated by AMH and Wnt signaling molecules (duct regression in males) and Hox genes (duct patterning in females). Most of the genes and molecular pathways known to be involved in Müllerian duct development have been elucidated through animal models, namely, the mouse and chicken. In addition, genetic analysis of humans with reproductive tract disorders has further defined molecular mechanisms of duct formation and differentiation. However, despite our current understanding of Müllerian duct development, some questions remain to be answered at the molecular genetic level. This article is categorized under: Early Embryonic Development > Development to the Basic Body Plan.


Subject(s)
Gene Expression Regulation, Developmental , Mullerian Ducts/embryology , Sex Differentiation , Animals , Cell Lineage , Female , Humans , LIM-Homeodomain Proteins/metabolism , Male , Mullerian Ducts/cytology , Mullerian Ducts/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...