1
|
Fu X, Liu Q, Song Y, Wang Y, Zhou Y, Wang S, Liu T, Zhuang F, Hu J, Sun Y. The molecule events expression of TGF-β/Smad signaling pathway in morphological and structural developmental characteristics of gonads in goose embryos. Poult Sci 2025; 104:104808. [PMID: 39823842 PMCID: PMC11786765 DOI: 10.1016/j.psj.2025.104808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
China is the largest producer and consumer of geese with significant social and economic value in agriculture. The Jilin White Goose, known for its excellent egg-laying and reproductive characteristics, is a prominent breeding breed in the northeast of China widely used for cross-breeding.Gonad development is a complex process, which will differentiate into testes or ovaries, thus affecting sex determination. Since the level of gonadal development in geese of different sexes is also related to their productivity, the study of gonadal function in geese is of great theoretical and practical importance. In this study, we used the embryonic gonads of Jilin white geese as the research object to reveal the morphologic and histologic developmental characteristics and process of the gonads of goose embryos from the 9th day to the 28th day and to investigate the mechanisms regulating the differentiation of the gonads. The results of ALP-PAS staining by histomorphological observations showed that the primordial germ cells in the female gonads were fully differentiated at embryonic day 19 (E19) and in the male gonads at embryonic day 18 (E18).Previous studies indicate that the TGF-β /Smad signaling pathway is crucial for sex differentiation and gamete formation in animals. In our study, we quantitatively analyzed key genes and proteins in this pathway within goose embryos. We found that TGF-βRI/II receptors and Smad2/3 are involved in gonadal development early in differentiation. Smad2 protein levels peaked at E18 in male gonads and E19 in female gonads, while Smad3 peaked at E10 for both. It has been suggested that there is a novel interaction between TGF-β and Wnt/β-catenin signaling pathway. In this study, the expression of LEF1, a protein downstream of the Wnt/β-catenin signaling pathway, peaked at E19 in the female gonads and at E18 in the male gonads, and the protein expression level of β-catenin was significantly increased in the female gonads at E19 and E28 and peaked at E28 in the male gonads. β-catenin expression levels were significantly higher in the male gonads than in the other periods and reached a minimum at E28. We injected the TMAO drug, which activates the TGF-β /Smad pathway, into goose embryos using the embryo injection technique to study how it affects gene and protein expression in this signaling pathway. We found that in female gonads, the levels of Smad2/3 proteins were lower than in the control group at E18, but higher than the control group at E19. In male gonads, the results for Smad3 proteins matched those of the females, while the results for Smad2 proteins were the opposite. This shows that TMAO activation of the TGF-β /Smad signaling pathway may impact gonadal development in goose embryos. In conclusion, this study suggest that the TGF-β /Smad and Wnt signaling pathways may enhance gonadal tissue development in goose embryos. The study identified key time points in gonadogenesis, providing a new theoretical basis for further research on the molecular mechanisms of gonadal development.
Collapse
Affiliation(s)
- Xianou Fu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiuyuan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yupu Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yudong Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuxuan Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Sihui Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Tuoya Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Fangming Zhuang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yongfeng Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Jilin Agricultural University, Ministry of Education, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
2
|
Kato S, Yokoyama T, Okunishi N, Narita H, Fujikawa T, Kirizuki Y, Mantani Y, Miki T, Hoshi N. Direct diffusion of anti-Müllerian hormone from both the cranial and caudal regions of the testis during early gonadal development in mice. Dev Dyn 2024; 253:296-311. [PMID: 37787412 DOI: 10.1002/dvdy.662] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/15/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND The Müllerian duct (MD), the primordium of the female reproductive tract, is also formed in males during the early stage of development, then regresses due to the anti-Müllerian hormone (AMH) secreted from the testes. However, the detailed diffusion pathway of AMH remains unclear. We herein investigated the mechanism by which AMH reaches the middle region of the MD using an organ culture system. RESULTS Injection of recombinant human AMH into the testis around the start of MD regression induced diffuse immunoreactivity in the mesonephros near the injection site. When the testis and mesonephros were cultured separately, the diameters of both cranial and middle MDs were significantly increased compared to the control. In the testis-mesonephros complex cultured by inhibiting the diffusion of AMH through the cranial region, the cranial MD diameter was significantly increased compared to the control, and there was no difference in middle MD diameter. CONCLUSIONS These results indicate that AMH, which infiltrates from the testis through the cranial region at physiological concentrations, induces regression of the cranial MD at the start of MD regression. They also indicate that AMH infiltrating through the caudal regions induces regression of the middle MD.
Collapse
Affiliation(s)
- Shiori Kato
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Toshifumi Yokoyama
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Nobusuke Okunishi
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Hiroto Narita
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Taisei Fujikawa
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Yusuke Kirizuki
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Youhei Mantani
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| | - Takanori Miki
- Faculty of Medicine, Departments of Anatomy and Neurobiology, Kagawa University, Miki-cho, Kagawa, Japan
| | - Nobuhiko Hoshi
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Japan
| |
Collapse
|
3
|
Tan JL, Major AT, Smith CA. Mini review: Asymmetric Müllerian duct development in the chicken embryo. Front Cell Dev Biol 2024; 12:1347711. [PMID: 38380340 PMCID: PMC10877723 DOI: 10.3389/fcell.2024.1347711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract. In humans, the Müllerian ducts differentiate into the Fallopian tubes, uterus and upper portion of the vagina. In birds and reptiles, the Müllerian ducts develop into homologous structures, the oviducts. The genetic and hormonal regulation of duct development is a model for understanding sexual differentiation. In males, the ducts typically undergo regression during embryonic life, under the influence of testis-derived Anti-Müllerian Hormone, AMH. In females, a lack of AMH during embryogenesis allows the ducts to differentiate into the female reproductive tract. In the chicken embryo, a long-standing model for development and sexual differentiation, Müllerian duct development in females in asymmetric. Only the left duct forms an oviduct, coincident with ovary formation only on the left side of the body. The right duct, together with the right gonad, becomes vestigial. The mechanism of this avian asymmetry has never been fully resolved, but is thought to involve local interplay between AMH and sex steroid hormones. This mini-review re-visits the topic, highlighting questions in the field and proposing a testable model for asymmetric duct development. We argue that current molecular and imaging techniques will shed new light on this curious asymmetry. Information on asymmetric duct development in the chicken model will inform our understanding of sexual differentiation in vertebrates more broadly.
Collapse
Affiliation(s)
| | | | - Craig A. Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
4
|
Nakajima T, Imai A, Ishii C, Tsuruyama K, Yamanaka R, Tomooka Y, Saito S, Adachi N, Kohno S, Sato T. SMAD2/3 signaling regulates initiation of mouse Wolffian ducts and proximal differentiation in Müllerian ducts. FEBS Open Bio 2024; 14:37-50. [PMID: 37953493 PMCID: PMC10761927 DOI: 10.1002/2211-5463.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023] Open
Abstract
Male and female reproductive tracts develop from anterior intermediate mesoderm with similar differentiation processes. The anterior intermediate mesoderm develops into the mesonephros, and the Wolffian duct initiates by epithelialization in the mesonephros. The Müllerian duct invaginates from the coelomic epithelium of the cranial mesonephros for ductal formation and is then regionalized into proximal to caudal female reproductive tracts. In this study, we focused on the epithelialization of the Wolffian duct, initiation of the Müllerian duct, and the regionalization step of the Müllerian ducts as a continuous process. By using intermediate mesodermal cells from mouse pluripotent stem cells, we identified that inhibition of SMAD2/3 signaling might be involved in the differentiation into mesenchymal cells, after which mesonephric cells might be then epithelialized during differentiation of the Wolffian duct. Aggregation of coelomic epithelial cells might be related to initiation of the Müllerian duct. Transcriptomic analysis predicted that consensus sequences of SMAD3/4 were enriched among highly expressed genes in the proximal Müllerian duct. SMAD2/3 signaling to regulate differentiation of the Wolffian duct was continuously activated in the proximal Müllerian duct and was involved in proximal and oviductal regionalization. Therefore, SMAD2/3 signaling may be finely tuned to regulate differentiation from initiation to regionalization steps.
Collapse
Affiliation(s)
- Tadaaki Nakajima
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Akihiro Imai
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Chihiro Ishii
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Kota Tsuruyama
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Risa Yamanaka
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Yasuhiro Tomooka
- Department of Biological Science and Technology, Faculty of Industrial Science and TechnologyTokyo University of ScienceJapan
| | - Shinta Saito
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Noritaka Adachi
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| | - Satomi Kohno
- Department of Biological SciencesSt. Cloud State UniversityMNUSA
| | - Tomomi Sato
- Department of ScienceYokohama City UniversityJapan
- Graduate School of NanobioscienceYokohama City UniversityJapan
| |
Collapse
|
5
|
Kanamori A, Kitani R, Oota A, Hirano K, Myosho T, Kobayashi T, Kawamura K, Kato N, Ansai S, Kinoshita M. Wnt4a Is Indispensable for Genital Duct Elongation but Not for Gonadal Sex Differentiation in the Medaka, Oryzias latipes. Zoolog Sci 2023; 40:348-359. [PMID: 37818883 DOI: 10.2108/zs230050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/24/2023] [Indexed: 10/13/2023]
Abstract
In most vertebrates, the oviducts and sperm ducts are derived from the Müllerian ducts and Wolffian ducts, respectively. However, in teleosts, the genital ducts are formed by the posterior extension of gonads in both sexes. Whether the genital ducts of teleosts are newly evolved organs or variants of Müllerian ducts is an important question for understanding evolutionary mechanisms of morphogenesis. One of the genes essential for Müllerian duct formation in mice is Wnt4, which is expressed in the mesenchyme and induces invagination of the coelomic epithelium and its posterior elongation. Here, we addressed the above question by examining genital duct development in mutants of two Wnt4 genes in the medaka (wnt4a is orthologous to mouse Wnt4, and wnt4b is paralogous). The wnt4b mutants had a short body but were fertile with normal genital ducts. In contrast, both male and female wnt4a mutants had their posterior elongation of the gonads stopped within or just outside the coelom. The mutants retained the posterior parts of ovarian cavities or sperm duct primordia, which are potential target tissues of Wnt4a. The gonads of female scl mutants (unable to synthesize sex steroids) lacked these tissues and did not develop genital ducts. Medaka wnt4a was expressed in the mesenchyme ventral to the genital ducts in both sexes. Taken together, the data strongly suggest that the mouse Müllerian ducts and the medaka genital ducts share homologous developmental processes. Additionally, the wnt4a or wnt4b single mutants and the double mutants did not show sex-reversal, implying that both genes are dispensable for gonadal sex differentiation in the medaka.
Collapse
Affiliation(s)
- Akira Kanamori
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan,
| | - Ryota Kitani
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Atsuko Oota
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Koudai Hirano
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Taijun Myosho
- Laboratory of Molecular Reproductive Biology, Institute for Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Tohru Kobayashi
- Laboratory of Molecular Reproductive Biology, Institute for Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Kouichi Kawamura
- Department of Marine Bioresources, Graduate School of Bioresources, Mie University, Mie 514-8507, Japan
| | - Naoyuki Kato
- Department of Environmental Science, Graduate School of Science and Technology, Niigata University, Niigata 950-2181, Japan
| | - Satoshi Ansai
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
- Laboratory of Bioresources, National Institute for Basic Biology, Aichi 445-8585, Japan
- Graduate School of Life Sciences, Tohoku University, Miyagi 980-8577, Japan
| | - Masato Kinoshita
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
6
|
Habiba M, Benagiano G, Guo SW. An Appraisal of the Tissue Injury and Repair (TIAR) Theory on the Pathogenesis of Endometriosis and Adenomyosis. Biomolecules 2023; 13:975. [PMID: 37371555 DOI: 10.3390/biom13060975] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
As understanding their pathogenesis remains elusive, both endometriosis and adenomyosis are often referred to as "enigmatic diseases". The uncertainty and heightened interest are reflected in the range of expressed views and opinions. There is a sense of urgency because of the entailed patient suffering. The plethora of opinions calls for a critical analysis of proposed theories, both old and new. A series of papers published since 2009 proposed that both endometriosis and adenomyosis originate from the same aberrations occurring within the uterus. This came to be recognized as the tissue injury and repair theory, and the newly coined term "archimetrosis" posits that the two diseases share the same origin. While the theory opens an interesting channel for exploration, its claim as a unifying theory necessitates a critical appraisal. We, thus, undertook this review of the theory and analyzed its underpinnings based on a comprehensive review of the literature. Our appraisal indicates that the theory is open to a range of criticisms. Chief among these is the need for confirmatory evidence of features of abnormal uterine contractility and the lack of data addressing the question of causality. In addition, the theory has, as yet, no supporting epidemiological evidence, which is a major weakness. The theory suffers as it is not open to the test of falsifiability, and it lacks the ability to make useful predictions. It has not addressed the questions, such as why only a small percentage of women develop adenomyosis or endometriosis, given the ubiquity of uterine peristalsis. On the other hand, the triggers and prevention of hyper- or dys-peristalsis become critical to a theory of causation. We conclude that additional supportive evidence is required for the theory to be accepted.
Collapse
Affiliation(s)
- Marwan Habiba
- Department of Health Sciences, University of Leicester and University Hospitals of Leicester, Leicester LE1 5WW, UK
| | - Giuseppe Benagiano
- Faculty of Medicine and Dentistry, Sapienza, University of Rome, 00161 Rome, Italy
| | - Sun-Wei Guo
- Research Institute, Shanghai Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai 200011, China
| |
Collapse
|
7
|
A re-appraisal of mesenchymal-epithelial transition (MET) in endometrial epithelial remodeling. Cell Tissue Res 2023; 391:393-408. [PMID: 36401092 PMCID: PMC9889438 DOI: 10.1007/s00441-022-03711-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
Abstract
Mesenchymal-epithelial transition (MET) is a mechanism of endometrial epithelial regeneration. It is also implicated in adenocarcinoma and endometriosis. Little is known about this process in normal uterine physiology. Previously, using pregnancy and menses-like mouse models, MET occurred only as an epithelial damage/repair mechanism. Here, we hypothesized that MET also occurs in other physiological endometrial remodeling events, outside of damage/repair, such as during the estrous cycle and adenogenesis (gland development). To investigate this, Amhr2-Cre-YFP/GFP mesenchyme-specific reporter mice were used to track the fate of mesenchymal-derived (MD) cells. Using EpCAM (epithelial marker), EpCAM+YFP+ MD-epithelial cells were identified in all stages of the estrous cycle except diestrus, in both postpartum and virgin mice. EpCAM+YFP+ MD-epithelial cells comprised up to 80% of the epithelia during estrogen-dominant proestrus and significantly declined to indistinguishable from control uteri in diestrus, suggesting MET is hormonally regulated. MD-epithelial cells were also identified during postnatal epithelial remodeling. MET occurred immediately after birth at postnatal day (P) 0.5 with EpCAM+GFP+ cells ranging from negligible (0.21%) to 82% of the epithelia. EpCAM+GFP+ MD-epithelial cells declined during initiation of adenogenesis (P8, avg. 1.75%) and then increased during gland morphogenesis (P14, avg. 10%). MD-epithelial cells expressed markers in common with non-MD-epithelial cells (e.g., EpCAM, FOXA2, ESR1, PGR). However, MD-epithelial cells were differentially regulated postnatally and in adults, suggesting a functional distinction in the two populations. We conclude that MET occurs not only as an epithelial damage/repair mechanism but also during other epithelial remodeling events, which to our knowledge has not been demonstrated in other tissues.
Collapse
|
8
|
Qin H, Wang J, Jia X, Zhi Y, Sun L, Zhang J, Wang J, Lu Y. Quantitative proteomics analysis of chicken embryos reveals key proteins that affect right gonadal degeneration in females. Proteomics 2022:e2200428. [PMID: 36574226 DOI: 10.1002/pmic.202200428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
In birds, embryonic gonads of females develop in a way different from mammals, with the left one develops into a functional ovary, while the right one degenerates during embryogenesis. Here, we examined the proteomics profiles of the female and male left and right gonads at embryonic day 6.5 (E6.5) with the label free tandem mass spectrometry proteomics technique. The relative protein abundance of the left and right gonads of female and male embryos was determined to identify their differential proteins. Overall, a total of 7726 proteins were identified, of which 79 and 54 proteins were significantly different in female and male right gonads compared with female left gonads and male left gonads respectively. Bioinformatics analysis showed that the proteins DMRT1, ZFPM2, TSHZ3 were potentially associated with the degeneration of the right gonads in female embryos. The proteomics in this study provide clues for further elucidation of the pathways of sex determination, sex differentiation, and right gonadal degeneration in birds.
Collapse
Affiliation(s)
- Haimei Qin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Reproductive Medicine, Guangxi Medical and health key discipline construction project of the Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Baise, China
| | - Jingyuan Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
- Zhang Jiagang Animal Husbandry and Veterinary Station, Zhang Jiagang, Jiangsu, China
| | - Xiaoxuan Jia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yifei Zhi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lingling Sun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiale Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Junli Wang
- Reproductive Medicine, Guangxi Medical and health key discipline construction project of the Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi Baise, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
9
|
Picard JY, Morin G, Devouassoux-Shisheboran M, Van der Smagt J, Klosowski S, Pienkowski C, Pierre-Renoult P, Masson C, Bole C, Josso N. Persistent Müllerian duct syndrome associated with genetic defects in the regulatory subunit of myosin phosphatase. Hum Reprod 2022; 37:2952-2959. [PMID: 36331510 DOI: 10.1093/humrep/deac239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
STUDY QUESTION Can mutations of genes other than AMH or AMHR2, namely PPP1R12A coding myosin phosphatase, lead to persistent Müllerian duct syndrome (PMDS)? SUMMARY ANSWER The detection of PPP1R12A truncation mutations in five cases of PMDS suggests that myosin phosphatase is involved in Müllerian regression, independently of the anti-Müllerian hormone (AMH) signaling cascade. WHAT IS KNOWN ALREADY Mutations of AMH and AMHR2 are detectable in an overwhelming majority of PMDS patients but in 10% of cases, both genes are apparently normal, suggesting that other genes may be involved. STUDY DESIGN, SIZE, DURATION DNA samples from 39 PMDS patients collected from 1990 to present, in which Sanger sequencing had failed to detect biallelic AMH or AMHR2 mutations, were screened by massive parallel sequencing. PARTICIPANTS/MATERIALS, SETTING, METHODS To rule out the possibility that AMH or AMHR2 mutations could have been missed, all DNA samples of good quality were analyzed by targeted next-generation sequencing. Twenty-four samples in which the absence of AMH or AMHR2 biallelic mutations was confirmed were subjected to whole-exome sequencing with the aim of detecting variants of other genes potentially involved in PMDS. MAIN RESULTS AND THE ROLE OF CHANCE Five patients out of 24 (21%) harbored deleterious truncation mutations of PP1R12A, the gene coding for the regulatory subunit of myosin phosphatase, were detected. In addition to PMDS, three of these patients presented with ileal and one with esophageal atresia. The congenital abnormalities associated with PMDS in our patients are consistent with those described in the literature for PPP1R12A variants and have never been described in cases of AMH or AMHR2 mutations. The role of chance is therefore extremely unlikely. LIMITATIONS, REASONS FOR CAUTION The main limitation of the study is the lack of experimental validation of the role of PPP1R12A in Müllerian regression. Only circumstantial evidence is available, myosin phosphatase is required for cell mobility, which plays a major role in Müllerian regression. Alternatively, PPP1R12A mutations could affect the AMH transduction pathway. WIDER IMPLICATIONS OF THE FINDINGS The study supports the conclusion that failure of Müllerian regression in males is not necessarily associated with a defect in AMH signaling. Extending the scope of molecular analysis should shed light upon the mechanism of the initial steps of male sex differentiation. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by la Fondation Maladies Rares, GenOmics 2021_0404 and la Fondation pour la Recherche Médicale, grant EQU201903007868. The authors report no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Jean-Yves Picard
- Sorbonne Université, INSERM, Centre de Recherches Saint-Antoine, Lipodystrophies, Adaptations Métaboliques et Hormonales et Vieillissement, UMR_S 938, Paris, France
| | - Gilles Morin
- Department of Medical Genetics, Centre Hospitalo-Universitaire d'Amiens, Amiens, France
| | | | | | - Serge Klosowski
- Service de Néonatologie, Centre Universitaire de Lens, Lens, France
| | | | | | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Descartes University, Sorbonne Paris Cite University, Paris, France
| | - Christine Bole
- Genomics Core Facility, Institut Imagine, Structure Fédérative de Recherche Necker, INSERM 1163, INSERM US24/CNRS UAR3633, Paris Descartes University, Sorbonne Paris Cité University, Paris, France
| | - Nathalie Josso
- Sorbonne Université, INSERM, Centre de Recherches Saint-Antoine, Lipodystrophies, Adaptations Métaboliques et Hormonales et Vieillissement, UMR_S 938, Paris, France
| |
Collapse
|
10
|
Yoshimatsu S, Kisu I, Qian E, Noce T. A New Horizon in Reproductive Research with Pluripotent Stem Cells: Successful In Vitro Gametogenesis in Rodents, Its Application to Large Animals, and Future In Vitro Reconstitution of Reproductive Organs Such as “Uteroid” and “Oviductoid”. BIOLOGY 2022; 11:biology11070987. [PMID: 36101367 PMCID: PMC9312112 DOI: 10.3390/biology11070987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Functional gametes, such as oocytes and spermatozoa, have been derived from rodent pluripotent stem cells, which can be applied to large animals and ultimately, to humans. In addition to summarizing these topics, we also review additional approaches for in vitro reconstitution of reproductive organs. This review illustrates intensive past efforts and future challenges on stem cell research for in vitro biogenesis in various mammalian models. Abstract Recent success in derivation of functional gametes (oocytes and spermatozoa) from pluripotent stem cells (PSCs) of rodents has made it feasible for future application to large animals including endangered species and to ultimately humans. Here, we summarize backgrounds and recent studies on in vitro gametogenesis from rodent PSCs, and similar approaches using PSCs from large animals, including livestock, nonhuman primates (NHPs), and humans. We also describe additional developing approaches for in vitro reconstitution of reproductive organs, such as the ovary (ovarioid), testis (testisoid), and future challenges in the uterus (uteroid) and oviduct (oviductoid), all of which may be derived from PSCs. Once established, these in vitro systems may serve as a robust platform for elucidating the pathology of infertility-related disorders and ectopic pregnancy, principle of reproduction, and artificial biogenesis. Therefore, these possibilities, especially when using human cells, require consideration of ethical issues, and international agreements and guidelines need to be raised before opening “Pandora’s Box”.
Collapse
Affiliation(s)
- Sho Yoshimatsu
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
- Research Fellow of Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Tokyo 102-0083, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-City 351-0198, Japan;
- Correspondence:
| | - Iori Kisu
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Emi Qian
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Toshiaki Noce
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-City 351-0198, Japan;
| |
Collapse
|
11
|
Machado DA, Ontiveros AE, Behringer RR. Mammalian uterine morphogenesis and variations. Curr Top Dev Biol 2022; 148:51-77. [DOI: 10.1016/bs.ctdb.2021.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Kapczuk K, Kędzia W. Primary Amenorrhea Due to Anatomical Abnormalities of the Reproductive Tract: Molecular Insight. Int J Mol Sci 2021; 22:ijms222111495. [PMID: 34768925 PMCID: PMC8584168 DOI: 10.3390/ijms222111495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/17/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Congenital anomalies of the female reproductive tract that present with primary amenorrhea involve Müllerian aplasia, also known as Mayer-Rokitansky-Küster-Hauser syndrome (MRKHS), and cervical and vaginal anomalies that completely obstruct the reproductive tract. Karyotype abnormalities do not exclude the diagnosis of MRKHS. Familial cases of Müllerian anomalies and associated malformations of the urinary and skeletal systems strongly suggest a complex genetic etiology, but so far, the molecular mechanism in the vast majority of cases remains unknown. Primary amenorrhea may also be the first presentation of complete androgen insensitivity syndrome, steroid 5α-reductase type 2 deficiency, 17β-hydroxysteroid dehydrogenase type 3 deficiency, and Leydig cells hypoplasia type 1; therefore, these disorders should be considered in the differential diagnosis of the congenital absence of the uterus and vagina. The molecular diagnosis in the majority of these cases can be established.
Collapse
|
13
|
Major AT, Estermann MA, Roly ZY, Smith CA. An evo-devo perspective of the female reproductive tract. Biol Reprod 2021; 106:9-23. [PMID: 34494091 DOI: 10.1093/biolre/ioab166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 01/22/2023] Open
Abstract
The vertebrate female reproductive tract has undergone considerable diversification over evolution, having become physiologically adapted to different reproductive strategies. This review considers the female reproductive tract from the perspective of evolutionary developmental biology (evo-devo). Very little is known about how the evolution of this organ system has been driven at the molecular level. In most vertebrates, the female reproductive tract develops from paired embryonic tubes, the Müllerian ducts. We propose that formation of the Müllerian duct is a conserved process that has involved co-option of genes and molecular pathways involved in tubulogenesis in the adjacent mesonephric kidney and Wolffian duct. Downstream of this conservation, genetic regulatory divergence has occurred, generating diversity in duct structure. Plasticity of the Hox gene code and wnt signaling, in particular, may underlie morphological variation of the uterus in mammals, and evolution of the vagina. This developmental plasticity in Hox and Wnt activity may also apply to other vertebrates, generating the morphological diversity of female reproductive tracts evident today.
Collapse
Affiliation(s)
- Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Zahida Y Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, 3800. Australia
| |
Collapse
|
14
|
Santana González L, Artibani M, Ahmed AA. Studying Müllerian duct anomalies - from cataloguing phenotypes to discovering causation. Dis Model Mech 2021; 14:269240. [PMID: 34160006 PMCID: PMC8246269 DOI: 10.1242/dmm.047977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Müllerian duct anomalies (MDAs) are developmental disorders of the Müllerian duct, the embryonic anlage of most of the female reproductive tract. The prevalence of MDAs is 6.7% in the general female population and 16.7% in women who exhibit recurrent miscarriages. Individuals affected by these anomalies suffer from high rates of infertility, first-trimester pregnancy losses, premature labour, placental retention, foetal growth retardation and foetal malpresentations. The aetiology of MDAs is complex and heterogeneous, displaying a range of clinical pictures that generally lack a direct genotype-phenotype correlation. De novo and familial cases sharing the same genomic lesions have been reported. The familial cases follow an autosomal-dominant inheritance, with reduced penetrance and variable expressivity. Furthermore, few genetic factors and molecular pathways underpinning Müllerian development and dysregulations causing MDAs have been identified. The current knowledge in this field predominantly derives from loss-of-function experiments in mouse and chicken models, as well as from human genetic association studies using traditional approaches, such as microarrays and Sanger sequencing, limiting the discovery of causal factors to few genetic entities from the coding genome. In this Review, we summarise the current state of the field, discuss limitations in the number of studies and patient samples that have stalled progress, and review how the development of new technologies provides a unique opportunity to overcome these limitations. Furthermore, we discuss how these new technologies can improve functional validation of potential causative alterations in MDAs. Summary: Here, we review the current knowledge about Müllerian duct anomalies in the context of new high-throughput technologies and model systems and their implications in the prevention of these disorders.
Collapse
Affiliation(s)
- Laura Santana González
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ahmed Ashour Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
15
|
Bergmann S, Schindler M, Munger C, Penfold CA, Boroviak TE. Building a stem cell-based primate uterus. Commun Biol 2021; 4:749. [PMID: 34140619 PMCID: PMC8211708 DOI: 10.1038/s42003-021-02233-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
The uterus is the organ for embryo implantation and fetal development. Most current models of the uterus are centred around capturing its function during later stages of pregnancy to increase the survival in pre-term births. However, in vitro models focusing on the uterine tissue itself would allow modelling of pathologies including endometriosis and uterine cancers, and open new avenues to investigate embryo implantation and human development. Motivated by these key questions, we discuss how stem cell-based uteri may be engineered from constituent cell parts, either as advanced self-organising cultures, or by controlled assembly through microfluidic and print-based technologies.
Collapse
Affiliation(s)
- Sophie Bergmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Magdalena Schindler
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Clara Munger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Christopher A Penfold
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
- Wellcome Trust - Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Cambridge, UK.
| | - Thorsten E Boroviak
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
| |
Collapse
|
16
|
Ghosh A, Syed SM, Kumar M, Carpenter TJ, Teixeira JM, Houairia N, Negi S, Tanwar PS. In Vivo Cell Fate Tracing Provides No Evidence for Mesenchymal to Epithelial Transition in Adult Fallopian Tube and Uterus. Cell Rep 2021; 31:107631. [PMID: 32402291 PMCID: PMC8094408 DOI: 10.1016/j.celrep.2020.107631] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
The mesenchymal to epithelial transition (MET) is thought to be involved in the maintenance, repair, and carcinogenesis of the fallopian tube (oviduct) and uterine epithelium. However, conclusive evidence for the conversion of mesenchymal cells to epithelial cells in these organs is lacking. Using embryonal cell lineage tracing with reporters driven by mesenchymal cell marker genes of the female reproductive tract (AMHR2, CSPG4, and PDGFRβ), we show that these reporters are also expressed by some oviductal and uterine epithelial cells at birth. These mesenchymal reporter-positive epithelial cells are maintained in adult mice across multiple pregnancies, respond to ovarian hormones, and form organoids. However, no labeled epithelial cells are present in any oviductal or uterine epithelia when mesenchymal cell labeling was induced in adult mice. Organoids developed from mice labeled in adulthood were also negative for mesenchymal reporters. Collectively, our work found no definitive evidence of MET in the adult fallopian tube and uterine epithelium. Mesenchymal to epithelial transition (MET) is postulated to be involved in the maintenance and regeneration of the epithelium of female reproductive organs. Here, Ghosh et al. report no definitive evidence of MET in the adult epithelium of oviduct and uterus using in vivo cell lineage tracing and organoids.
Collapse
Affiliation(s)
- Arnab Ghosh
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Shafiq M Syed
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Manish Kumar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Tyler J Carpenter
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Nathaniel Houairia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Sumedha Negi
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Pradeep S Tanwar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
17
|
The History of the Discovery of Ectopic Epithelial Cells in Lower Peritoneal Organs: The So-Called Mucosal Invasion. REPRODUCTIVE MEDICINE 2021. [DOI: 10.3390/reprodmed2020008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Through microscopy, early researchers identified the epithelium on the inner surfaces of the uterus, cervix and Fallopian tubes. The identification of ectopic epithelium was gradual, starting from the gross pathology study of unusual cystic lesions. Towards the end of the nineteenth century, attention focused on the epithelium as a critical component. The term ‘adenomyoma’ was coined around eighteen eighty to designate the majority of mucosa-containing lesions. Several theories were advanced to explain its aetiology. In the main, lesions were considered to arise from invasion from uterine epithelium; implantation of endometrium through retrograde menstruation; hematogenous or lymphatic spread; or from embryonic remnants. Although initially widely rejected, around 1920, an almost unanimous consensus formed on the endometrial nature of epithelial invasions. During the following years, adenomyosis and endometriosis came to be used to distinguished lesions within or outside the uterus. Adenomyosis was attributed to direct infiltration of uterine mucosa into the myometrium, and endometriosis to the implantation of endometrial cells and stroma into the peritoneal cavity through retrograde menstruation. Around the same time, ovarian lesions, initially described as ovarian hematomas or chocolate cysts, were regarded as a form of endometriosis. Three variants of endometriosis were thus described: superficial peritoneal, deep nodular and ovarian endometriomas. Ectopic epithelium has long been recognised as having similarities to tubal, or cervical epithelium. Lesions containing mixed epithelium are often termed Müllerianosis. This article demonstrates the stepwise evolution of knowledge, the role of the pioneers and the difficulties that needed to be overcome. It also demonstrates the value of collaboration and the inter-connected nature of the scientific endeavour.
Collapse
|
18
|
Santana Gonzalez L, Rota IA, Artibani M, Morotti M, Hu Z, Wietek N, Alsaadi A, Albukhari A, Sauka-Spengler T, Ahmed AA. Mechanistic Drivers of Müllerian Duct Development and Differentiation Into the Oviduct. Front Cell Dev Biol 2021; 9:605301. [PMID: 33763415 PMCID: PMC7982813 DOI: 10.3389/fcell.2021.605301] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
The conduits of life; the animal oviducts and human fallopian tubes are of paramount importance for reproduction in amniotes. They connect the ovary with the uterus and are essential for fertility. They provide the appropriate environment for gamete maintenance, fertilization and preimplantation embryonic development. However, serious pathologies, such as ectopic pregnancy, malignancy and severe infections, occur in the oviducts. They can have drastic effects on fertility, and some are life-threatening. Despite the crucial importance of the oviducts in life, relatively little is known about the molecular drivers underpinning the embryonic development of their precursor structures, the Müllerian ducts, and their successive differentiation and maturation. The Müllerian ducts are simple rudimentary tubes comprised of an epithelial lumen surrounded by a mesenchymal layer. They differentiate into most of the adult female reproductive tract (FRT). The earliest sign of Müllerian duct formation is the thickening of the anterior mesonephric coelomic epithelium to form a placode of two distinct progenitor cells. It is proposed that one subset of progenitor cells undergoes partial epithelial-mesenchymal transition (pEMT), differentiating into immature Müllerian luminal cells, and another subset undergoes complete EMT to become Müllerian mesenchymal cells. These cells invaginate and proliferate forming the Müllerian ducts. Subsequently, pEMT would be reversed to generate differentiated epithelial cells lining the fully formed Müllerian lumen. The anterior Müllerian epithelial cells further specialize into the oviduct epithelial subtypes. This review highlights the key established molecular and genetic determinants of the processes involved in Müllerian duct development and the differentiation of its upper segment into oviducts. Furthermore, an extensive genome-wide survey of mouse knockout lines displaying Müllerian or oviduct phenotypes was undertaken. In addition to widely established genetic determinants of Müllerian duct development, our search has identified surprising associations between loss-of-function of several genes and high-penetrance abnormalities in the Müllerian duct and/or oviducts. Remarkably, these associations have not been investigated in any detail. Finally, we discuss future directions for research on Müllerian duct development and oviducts.
Collapse
Affiliation(s)
- Laura Santana Gonzalez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ioanna A Rota
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Developmental Immunology Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matteo Morotti
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Zhiyuan Hu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Nina Wietek
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Abdulkhaliq Alsaadi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ashwag Albukhari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ahmed A Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Habiba M, Heyn R, Bianchi P, Brosens I, Benagiano G. The development of the human uterus: morphogenesis to menarche. Hum Reprod Update 2020; 27:1-26. [PMID: 33395479 DOI: 10.1093/humupd/dmaa036] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
There is emerging evidence that early uterine development in humans is an important determinant of conditions such as ontogenetic progesterone resistance, menstrual preconditioning, defective deep placentation and pre-eclampsia in young adolescents. A key observation is the relative infrequency of neonatal uterine bleeding and hormone withdrawal at birth. The origin of the uterus from the fusion of the two paramesonephric, or Müllerian, ducts was described almost 200 years ago. The uterus forms around the 10th week of foetal life. The uterine corpus and the cervix react differently to the circulating steroid hormones during pregnancy. Adult uterine proportions are not attained until after puberty. It is unclear if the endometrial microbiome and immune response-which are areas of growing interest in the adult-play a role in the early stages of uterine development. The aim is to review the phases of uterine development up until the onset of puberty in order to trace the origin of abnormal development and to assess current knowledge for features that may be linked to conditions encountered later in life. The narrative review incorporates literature searches of Medline, PubMed and Scopus using the broad terms individually and then in combination: uterus, development, anatomy, microscopy, embryology, foetus, (pre)-puberty, menarche, microbiome and immune cells. Identified articles were assessed manually for relevance, any linked articles and historical textbooks. We included some animal studies of molecular mechanisms. There are competing theories about the contributions of the Müllerian and Wolffian ducts to the developing uterus. Endometrium features are suggestive of an oestrogen effect at 16-20 weeks gestation. The discrepancy in the reported expression of oestrogen receptor is likely to be related to the higher sensitivity of more recent techniques. Primitive endometrial glands appear around 20 weeks. Features of progestogen action are expressed late in the third trimester. Interestingly, progesterone receptor expression is higher at mid-gestation than at birth when features of endometrial maturation are rare. Neonatal uterine bleeding occurs in around 5% of neonates. Myometrial differentiation progresses from the mesenchyme surrounding the endometrium at the level of the cervix. During infancy, the uterus and endometrium remain inactive. The beginning of uterine growth precedes the onset of puberty and continues for several years after menarche. Uterine anomalies may result from fusion defects or atresia of one or both Müllerian ducts. Organogenetic differentiation of Müllerian epithelium to form the endometrial and endocervical epithelium may be independent of circulating steroids. A number of genes have been identified that are involved in endometrial and myometrial differentiation although gene mutations have not been demonstrated to be common in cases of uterine malformation. The role, if any, of the microbiome in relation to uterine development remains speculative. Modern molecular techniques applied to rodent models have enhanced our understanding of uterine molecular mechanisms and their interactions. However, little is known about functional correlates or features with relevance to adult onset of uterine disease in humans. Prepubertal growth and development lends itself to non-invasive diagnostics such as ultrasound and MRI. Increased awareness of the occurrence of neonatal uterine bleeding and of the potential impact on adult onset disease may stimulate renewed research in this area.
Collapse
Affiliation(s)
- Marwan Habiba
- Department of Health Sciences, University of Leicester and University Hospitals of Leicester, Leicester Royal Infirmary, Leicester, UK
| | - Rosemarie Heyn
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Paola Bianchi
- Department of Medico-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Ivo Brosens
- Faculty of Medicine, Catholic University of Leuven, Leuven, Belgium
| | - Giuseppe Benagiano
- Department of Maternal and Child Health, Gynaecology and Urology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
20
|
Roly ZY, Godini R, Estermann MA, Major AT, Pocock R, Smith CA. Transcriptional landscape of the embryonic chicken Müllerian duct. BMC Genomics 2020; 21:688. [PMID: 33008304 PMCID: PMC7532620 DOI: 10.1186/s12864-020-07106-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
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-β 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.
Collapse
Affiliation(s)
- Zahida Yesmin Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Rasoul Godini
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Roger Pocock
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.
| |
Collapse
|
21
|
Hamanaka K, Takata A, Uchiyama Y, Miyatake S, Miyake N, Mitsuhashi S, Iwama K, Fujita A, Imagawa E, Alkanaq AN, Koshimizu E, Azuma Y, Nakashima M, Mizuguchi T, Saitsu H, Wada Y, Minami S, Katoh-Fukui Y, Masunaga Y, Fukami M, Hasegawa T, Ogata T, Matsumoto N. MYRF haploinsufficiency causes 46,XY and 46,XX disorders of sex development: bioinformatics consideration. Hum Mol Genet 2020; 28:2319-2329. [PMID: 30985895 DOI: 10.1093/hmg/ddz066] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/06/2019] [Accepted: 03/21/2019] [Indexed: 12/30/2022] Open
Abstract
Disorders of sex development (DSDs) are defined as congenital conditions in which chromosomal, gonadal or anatomical sex is atypical. In many DSD cases, genetic causes remain to be elucidated. Here, we performed a case-control exome sequencing study comparing gene-based burdens of rare damaging variants between 26 DSD cases and 2625 controls. We found exome-wide significant enrichment of rare heterozygous truncating variants in the MYRF gene encoding myelin regulatory factor, a transcription factor essential for oligodendrocyte development. All three variants occurred de novo. We identified an additional 46,XY DSD case of a de novo damaging missense variant in an independent cohort. The clinical symptoms included hypoplasia of Müllerian derivatives and ovaries in 46,XX DSD patients, defective development of Sertoli and Leydig cells in 46,XY DSD patients and congenital diaphragmatic hernia in one 46,XY DSD patient. As all of these cells and tissues are or partly consist of coelomic epithelium (CE)-derived cells (CEDC) and CEDC developed from CE via proliferaiton and migration, MYRF might be related to these processes. Consistent with this hypothesis, single-cell RNA sequencing of foetal gonads revealed high expression of MYRF in CE and CEDC. Reanalysis of public chromatin immunoprecipitation sequencing data for rat Myrf showed that genes regulating proliferation and migration were enriched among putative target genes of Myrf. These results suggested that MYRF is a novel causative gene of 46,XY and 46,XX DSD and MYRF is a transcription factor regulating CD and/or CEDC proliferation and migration, which is essential for development of multiple organs.
Collapse
Affiliation(s)
| | | | - Yuri Uchiyama
- Department of Human Genetics.,Department of Oncology
| | - Satoko Miyatake
- Department of Human Genetics.,Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | | | | | | | | | | | | | | | - Yoshiki Azuma
- Department of Human Genetics.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | | | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yuka Wada
- Department of Neonatology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Sawako Minami
- Deparment of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Yuko Katoh-Fukui
- Department of Molecular Endocrinology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Yohei Masunaga
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | | |
Collapse
|
22
|
Guioli S, Zhao D, Nandi S, Clinton M, Lovell-Badge R. Oestrogen in the chick embryo can induce chromosomally male ZZ left gonad epithelial cells to form an ovarian cortex that can support oogenesis. Development 2020; 147:dev181693. [PMID: 32001442 PMCID: PMC7055392 DOI: 10.1242/dev.181693] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/16/2020] [Indexed: 12/25/2022]
Abstract
In chickens, the embryonic ovary differentiates into two distinct domains before meiosis: a steroidogenic core (the female medulla), overlain by the germ cell niche (the cortex). The differentiation of the medulla is a cell-autonomous process based on chromosomal sex identity (CASI). In order to address the extent to which cortex differentiation depends on intrinsic or extrinsic factors, we generated models of gonadal intersex by mixing ZW (female) and ZZ (male) cells in gonadal chimeras, or by altering oestrogen levels of ZW and ZZ embryos. We found that CASI does not apply to the embryonic cortex. Both ZW and ZZ cells can form the cortex and this can happen independently of the phenotypic sex of the medulla as long as oestrogen is provided. We also show that the cortex-promoting activity of oestrogen signalling is mediated via estrogen receptor alpha within the left gonad epithelium. However, the presence of a medulla with an 'intersex' or male phenotype may compromise germ cell progression into meiosis, causing cortical germ cells to remain in an immature state in the embryo.
Collapse
Affiliation(s)
| | - Debiao Zhao
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Sunil Nandi
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Michael Clinton
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | | |
Collapse
|
23
|
Seishima R, Leung C, Yada S, Murad KBA, Tan LT, Hajamohideen A, Tan SH, Itoh H, Murakami K, Ishida Y, Nakamizo S, Yoshikawa Y, Wong E, Barker N. Neonatal Wnt-dependent Lgr5 positive stem cells are essential for uterine gland development. Nat Commun 2019; 10:5378. [PMID: 31772170 PMCID: PMC6879518 DOI: 10.1038/s41467-019-13363-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Wnt signaling is critical for directing epithelial gland development within the uterine lining to ensure successful gestation in adults. Wnt-dependent, Lgr5-expressing stem/progenitor cells are essential for the development of glandular epithelia in the intestine and stomach, but their existence in the developing reproductive tract has not been investigated. Here, we employ Lgr5-2A-EGFP/CreERT2/DTR mouse models to identify Lgr5-expressing cells in the developing uterus and to evaluate their stem cell identity and function. Lgr5 is broadly expressed in the uterine epithelium during embryogenesis, but becomes largely restricted to the tips of developing glands after birth. In-vivo lineage tracing/ablation/organoid culture assays identify these gland-resident Lgr5high cells as Wnt-dependent stem cells responsible for uterine gland development. Adjacent Lgr5neg epithelial cells within the neonatal glands function as essential niche components to support the function of Lgr5high stem cells ex-vivo. These findings constitute a major advance in our understanding of uterine development and lay the foundations for investigating potential contributions of Lgr5+ stem/progenitor cells to uterine disorders. Uterine gland development is essential for successful embryo implantation, decidua formation and placental development. Here the authors demonstrate that neonatal Wnt-dependent Lgr5 expressing stem/progenitor cells at the tips of developing glands are indispensable for uterine gland development.
Collapse
Affiliation(s)
- Ryo Seishima
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | - Carly Leung
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | - Swathi Yada
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | | | - Liang Thing Tan
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | | | - Si Hui Tan
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | - Hideki Itoh
- A*STAR Skin Research Institute of Singapore, Singapore, 138648, Singapore
| | - Kazuhiro Murakami
- Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yoshihiro Ishida
- Department of Dermatology, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, 606-8501, Japan
| | - Satoshi Nakamizo
- A*STAR Skin Research Institute of Singapore, Singapore, 138648, Singapore
| | - Yusuke Yoshikawa
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | - Esther Wong
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore, 138648, Singapore. .,Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan. .,School of Biological Sciences, Nanyang Technological University, Singapore, 308232, Singapore.
| |
Collapse
|
24
|
Franke-Radowiecka A, Prozorowska E, Zalecki M, Jackowiak H, Kaleczyc J. Innervation of internal female genital organs in the pig during prenatal development. J Anat 2019; 235:1007-1017. [PMID: 31347705 DOI: 10.1111/joa.13052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2019] [Indexed: 11/30/2022] Open
Abstract
This study investigated the innervation of internal genital organs in 5-, 7- and 10-week-old female pig foetuses using single and double-labelling immunofluorescence methods. The structure and topography of the organs was examined using a scanning electron microscope (SEM). The investigations revealed differences in the innervation between the three developmental periods. Immunostaining for protein gene product 9.5 (PGP; general neural marker) disclosed solitary nerve fibres in the external part of the gonadal ridge and just outside of the mesenchyme surrounding mesonephric ducts in 5-week-old foetuses. Double-labelling immunohistochemistry revealed that nerve fibres associated with the ridge expressed dopamine β-hydroxylase (DβH; adrenergic marker) or vesicular acetylcholine transporter (VAChT; cholinergic marker). In 7-week-old foetuses, the PGP-positive nerve terminals were absent from the gonad but some of them ran outside and along, and sometimes penetrated into the mesenchyme surrounding the tubal and uterine segments of the paramesonephric ducts and uterovaginal canal. Few axons penetrated into the mesenchyme. DβH-positive fibres were found in single nerve strands or bundles distributed at the edge of the mesenchyme. VAChT-positive nerve terminals formed delicate bundles located at the edge of the mesenchyme, and the single nerves penetrated into the mesenchyme. DβH was also expressed by neurons which formed cell clusters comprising also DβH- or VAChT-positive nerve fibres. In 10-week-old foetuses, PGP-positive nerve fibres were still absent from the ovary but some were distributed in the mesenchyme associated with the uterovaginal canal and uterine and a tubal segment of the paramesonephric ducts, respectively. DβH- or VAChT-positive nerve fibres were distributed at the periphery of the mesenchyme associated with the uterovaginal canal. Some DβH- and many VAChT-positive nerve fibres were evenly distributed throughout the mesenchyme. The clusters of nerve cells comprised DβH-positive perikarya and DβH- or VAChT-positive nerve fibres. The investigations revealed no DβH/VAChT-positive nerve fibres or neurons as well as no nerve structures stained for calcitonin gene-related peptide and/or substance P (sensory markers) associated with the genital organs in the studied prenatal periods.
Collapse
Affiliation(s)
- Amelia Franke-Radowiecka
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Ewelina Prozorowska
- Department of Histology and Embryology, Poznań University of Life Sciences, Poznań, Poland
| | - Michal Zalecki
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Hanna Jackowiak
- Department of Histology and Embryology, Poznań University of Life Sciences, Poznań, Poland
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
25
|
Alsafy MAM, El-Gendy SAA, Karkoura AA, Naguib D. Light and Scanning Electron Microscopic Examination of the Chicken Oviduct during the Embryonic and Posthatching Stages. J Microsc Ultrastruct 2019; 7:9-13. [PMID: 31008051 PMCID: PMC6442323 DOI: 10.4103/jmau.jmau_12_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
The present study aimed to study the sequence of developing the oviduct of the Alexandria chicken during the embryonic and posthatching period by using the light and scanning electron microscopic (SEM) examination. The Mullerian duct began to appear as left and right urogenital ridges composed of stratified cuboidal epithelium at the ventrolateral aspect of the mesonephros at the 5-day-old embryo. At the 6-day-old embryo, the left urogenital ridge canalized and the tubal wall surrounded a circular lumen composed of three cellular components; inner simple columnar epithelium, multilayers of mesenchymal cells, and outer stratified cuboidal epithelium. At the 8-day-old embryo, the inner tubal layer became composed of simple-to-pseudostratified ciliated columnar epithelium, the density of the mesenchymal cells increased, and the outer layer became simple squamous epithelium at the medial aspect of the duct and stratified epithelium at the lateral aspect of the duct. The left oviduct of the 1-day-old chick resembled the oviduct of 8-day-old embryo except the SEM observations of the tunica mucosa of the 1-day-old chick which showed extensive mucosal folds with many straight cilia. At the 1-week-old chick, the left oviduct showed a folded lumen surrounded by simple columnar ciliated epithelial layer followed by a layer of mesenchymal cells, many layers of smooth muscles surrounded the mesenchymal cells layer and outer simple squamous epithelium layer. At the 1-month-old chick, the left oviduct wall was composed of five layers surrounded by a star-shaped lumen.
Collapse
Affiliation(s)
- Mohamed A M Alsafy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Samir A A El-Gendy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Ashraf A Karkoura
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Doha Naguib
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
26
|
Generation of Progesterone-Responsive Endometrial Stromal Fibroblasts from Human Induced Pluripotent Stem Cells: Role of the WNT/CTNNB1 Pathway. Stem Cell Reports 2018; 11:1136-1155. [PMID: 30392973 PMCID: PMC6234962 DOI: 10.1016/j.stemcr.2018.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/26/2022] Open
Abstract
Defective endometrial stromal fibroblasts (EMSFs) contribute to uterine factor infertility, endometriosis, and endometrial cancer. Induced pluripotent stem cells (iPSCs) derived from skin or bone marrow biopsies provide a patient-specific source that can be differentiated to various cells types. Replacement of abnormal EMSFs is a potential novel therapeutic approach for endometrial disease; however, the methodology or mechanism for differentiating iPSCs to EMSFs is unknown. The uterus differentiates from the intermediate mesoderm (IM) to form coelomic epithelium (CE) followed by the Müllerian duct (MD). Here, we successfully directed the differentiation of human iPSCs (hiPSCs) through IM, CE, and MD to EMSFs under molecularly defined embryoid body culture conditions using specific hormonal treatments. Activation of CTNNB1 was essential for expression of progesterone receptor that mediated the final differentiation step of EMSFs before implantation. These hiPSC-derived tissues illustrate the potential for iPSC-based endometrial regeneration for future cell-based treatments. We developed a molecularly defined system for differentiating hiPSCs to EMSFs hiPSC-derived EMSFs undergo decidualization in response to hormonal stimulation D14 embryoid bodies recapitulate the molecular signature of primary EMSFs The WNT/CTNNB1 pathway is required for induction of EMSF from hiPSCs
Collapse
|
27
|
Mesothelial-mesenchymal transitions in embryogenesis. Semin Cell Dev Biol 2018; 92:37-44. [PMID: 30243860 DOI: 10.1016/j.semcdb.2018.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 09/03/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
Most animals develop coelomic cavities lined by an epithelial cell layer called the mesothelium. Embryonic mesothelial cells have the ability to transform into mesenchymal cells which populate many developing organs contributing to their connective and vascular tissues, and also to organ-specific cell types. Furthermore, embryonic mesothelium and mesothelial-derived cells produce essential signals for visceral morphogenesis. We review the most relevant literature about the mechanisms regulating the embryonic mesothelial-mesenchymal transition, the developmental fate of the mesothelial-derived cells and other functions of the embryonic mesothelium, such as its contribution to the establishment of left-right visceral asymmetries or its role in limb morphogenesis.
Collapse
|
28
|
Yamamoto A, Omotehara T, Miura Y, Takada T, Yoneda N, Hirano T, Mantani Y, Kitagawa H, Yokoyama T, Hoshi N. The mechanisms underlying the effects of AMH on Müllerian duct regression in male mice. J Vet Med Sci 2018. [PMID: 29526868 PMCID: PMC5938179 DOI: 10.1292/jvms.18-0023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anti-Müllerian hormone (AMH) produced in the developing testis induces the regression of the Müllerian duct, which develops into the oviducts, uterus and upper vagina. In our true hermaphrodite mouse with an ovary on one side and a testis on the other (O/T), the oviduct and uterus are present only on the ovary side, and nothing derived from the Müllerian duct is present on the testis side. Here, we investigate the mechanism underlying the unilateral Müllerian duct regression and the mode of AMH signaling, by performing immunohistology, Western blotting, and organ culture analyses. The histological analysis revealed that during the start of the Müllerian duct regression, the duct in the O/T mice was clearly regressed on the AMH-positive testis side compared to the AMH-negative ovary side. The immunohistochemistry showed a diffuse immunoreaction of AMH in the interstitium surrounding the testis cord and boundary region between the testis and mesonephros, especially in the cranial portion. Western blotting revealed that the amount of AMH in the cranial half of the mesonephros was larger than that in the caudal half. AMH injected into the gonads in organ culture induced the regression of the Müllerian duct via the interstitium of the organ. These results suggest that AMH acts on the Müllerian duct in male mice by exuding into the interstitium surrounding the testis cord and infiltrating through the cranial region from the testis to the mesonephros.
Collapse
Affiliation(s)
- Anzu Yamamoto
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Takuya Omotehara
- Department of Anatomy, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo 160-8402, Japan
| | - Yuuka Miura
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Tadashi Takada
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Naoki Yoneda
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Tetsushi Hirano
- Division of Drug and Structural Research, Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Youhei Mantani
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Hiroshi Kitagawa
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Toshifumi Yokoyama
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| | - Nobuhiko Hoshi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada, Kobe, Hyogo 657-8501, Japan
| |
Collapse
|
29
|
Roly ZY, Backhouse B, Cutting A, Tan TY, Sinclair AH, Ayers KL, Major AT, Smith CA. The cell biology and molecular genetics of Müllerian duct development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e310. [DOI: 10.1002/wdev.310] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 11/06/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Zahida Yesmin Roly
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| | - Brendan Backhouse
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew Cutting
- Biology Laboratory, Faculty of ScienceThe University of MelbourneMelbourneVictoriaAustralia
| | - Tiong Yang Tan
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew H. Sinclair
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Katie L. Ayers
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalMelbourneVictoriaAustralia
| | - Andrew T. Major
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| | - Craig A. Smith
- Monash Biomedicine Discovery Institute, Department of Anatomy and Development BiologyMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
30
|
Molecular mechanisms of development of the human fetal female reproductive tract. Differentiation 2017; 97:54-72. [PMID: 29053991 DOI: 10.1016/j.diff.2017.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 01/30/2023]
Abstract
Human female reproductive tract development rests mostly upon hematoxilyn and eosin stained sections despite recent advances on molecular mechanisms in mouse studies. We report application of immunohistochemical methods to explore the ontogeny of epithelial and mesenchymal differentiation markers (keratins, homobox proteins, steroid receptors), transcription factors and signaling molecules (TP63 and RUNX1) during human female reproductive tract development. Keratins 6, 7, 8, 10, 14 and 19 (KRT6, KRT7, KRT8, KRT10, KRT14, KRT19) were expressed in a temporally and spatially dynamic fashion. The undifferentiated Müllerian duct and uterovaginal canal, lined by simple columnar epithelia, expressed KRT7, KRT8 and KRT19. Glandular derivatives of the Müllerian duct (uterine tube, uterine corpus and endocervix) maintained expression of these keratins, while tissues that undergo stratified squamous differentiation (exocervix and vagina) expressed KRT6, KRT14 and KRT10 during development in an age-dependent fashion. TP63 and RUNX1 were expressed prior to KRT14, as these two transcription factors are known to be upstream from KRT14 in developing Müllerian epithelium. In the vagina, KRT10, a marker of terminal differentiation, appeared after endogenous estrogens transformed the epithelium to a thick glycogenated squamous epithelium. Uroplakin, a protein unique to urothelium, was expressed only in the bladder, urethra and vaginal introitus, but not in the female reproductive tract itself. Mesenchymal differentiation was examined through immunostaining for HOXA11 (expressed in uterine mesenchyme) and ISL1 (expressed in vaginal mesenchyme). A detailed ontogeny of estrogen receptor alpha (ESR1), progesterone receptor (PGR) and the androgen receptor (AR) provides the mechanistic underpinning for the teratogenicity of estrogens, progestins and androgens on female reproductive tract development. Immunohistochemical analysis of differentiation markers and signaling molecules advance our understanding of normal development of the human female reproductive tract. These observations demonstrate remarkable similarities in mouse and human female reproductive tract development, but also highlight some key differences.
Collapse
|
31
|
Hirst CE, Serralbo O, Ayers KL, Roeszler KN, Smith CA. Genetic Manipulation of the Avian Urogenital System Using In Ovo Electroporation. Methods Mol Biol 2017; 1650:177-190. [PMID: 28809021 DOI: 10.1007/978-1-4939-7216-6_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
One of the advantages of the avian embryo as an experimental model is its in ovo development and hence accessibility for genetic manipulation. Electroporation has been used extensively in the past to study gene function in chicken and quail embryos . Readily accessible tissues such as the neural tube, somites, and limb bud, in particular, have been targeted. However, more inaccessible tissues, such as the embryonic urogenital system , have proven more challenging to study. Here, we describe the use of in ovo electroporation of TOL2 vectors or RCASBP avian viral vectors for the rapid functional analysis of genes involved in avian sex determination and urogenital development . In the context of the developing urogenital system , these vectors have inherent advantages and disadvantages, which will be considered here. Either vector can both be used for mis-expressing a gene and for targeting endogenous gene knockdown via expression of short hairpin RNAs (shRNAs). Both of these vectors integrate into the genome and are hence spread throughout developing tissues. Going forward, electroporation could be combined with CRISPR/Cas9 technology for targeted genome editing in the avian urogenital system .
Collapse
Affiliation(s)
- Claire E Hirst
- Department of Anatomy and Development Biology, Monash University, Clayton, VIC, 3800, USA
| | - Olivier Serralbo
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, USA
| | - Katie L Ayers
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, Melbourne, VIC, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Kelly N Roeszler
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, Melbourne, VIC, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Craig A Smith
- Department of Anatomy and Development Biology, Monash University, Clayton, VIC, 3800, USA.
| |
Collapse
|
32
|
Atsuta Y, Takahashi Y. Early formation of the Müllerian duct is regulated by sequential actions of BMP/Pax2 and FGF/Lim1 signaling. Development 2016; 143:3549-3559. [PMID: 27578782 DOI: 10.1242/dev.137067] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022]
Abstract
The Müllerian duct (MD) and Wolffian duct (WD) are embryonic tubular tissues giving rise to female and male reproductive tracts, respectively. In amniote embryos, both MD and WD emerge in both sexes, but subsequently degenerate in the males and females, respectively. Here, by using MD-specific gene manipulations in chicken embryos, we identify the molecular and cellular mechanisms that link early MD specification to tubular invagination. Early (pre-)specification of MD precursors in the coelomic epithelium requires BMP signaling and its downstream target Pax2 in a WD-independent process. Subsequently, the BMP/Pax2 axis induces Lim1 expression, a hallmark of MD specification, for which FGF/ERK and WD-derived signals are also required. Finally, the sequential actions of the BMP/Pax2 and FGF/Lim1 axes culminate in epithelial invagination to form a tubular structure driven by an apical constriction, where apical accumulation of phospho-myosin light chain is positively regulated by FGF/ERK signaling. Our study delineates mechanisms governing the early formation of the MD, and also serves as a model of how an epithelial cell sheet is transformed to a tubular structure, a process seen in a variety of developmental contexts.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan AMED Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
33
|
Jacquinet A, Millar D, Lehman A. Etiologies of uterine malformations. Am J Med Genet A 2016; 170:2141-72. [PMID: 27273803 DOI: 10.1002/ajmg.a.37775] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 03/10/2016] [Indexed: 12/11/2022]
Abstract
Ranging from aplastic uterus (including Mayer-Rokitansky-Kuster-Hauser syndrome) to incomplete septate uterus, uterine malformations as a group are relatively frequent in the general population. Specific causes remain largely unknown. Although most occurrences ostensibly seem sporadic, familial recurrences have been observed, which strongly implicate genetic factors. Through the study of animal models, human syndromes, and structural chromosomal variation, several candidate genes have been proposed and subsequently tested with targeted methods in series of individuals with isolated, non-isolated, or syndromic uterine malformations. To date, a few genes have garnered strong evidence of causality, mainly in syndromic presentations (HNF1B, WNT4, WNT7A, HOXA13). Sequencing of candidate genes in series of individuals with isolated uterine abnormalities has been able to suggest an association for several genes, but confirmation of a strong causative effect is still lacking for the majority of them. We review the current state of knowledge about the developmental origins of uterine malformations, with a focus on the genetic variants that have been implicated or associated with these conditions in humans, and we discuss potential reasons for the high rate of negative results. The evidence for various environmental and epigenetic factors is also reviewed. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Adeline Jacquinet
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Center for Human Genetics, Centre Hospitalier Universitaire and University of Liège, Liège, Belgium
| | - Debra Millar
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, Canada
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Child and Family Research Institute, Vancouver, Canada
| |
Collapse
|
34
|
Prunskaite-Hyyryläinen R, Skovorodkin I, Xu Q, Miinalainen I, Shan J, Vainio SJ. Wnt4 coordinates directional cell migration and extension of the Müllerian duct essential for ontogenesis of the female reproductive tract. Hum Mol Genet 2015; 25:1059-73. [PMID: 26721931 PMCID: PMC4764189 DOI: 10.1093/hmg/ddv621] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 12/21/2015] [Indexed: 12/27/2022] Open
Abstract
The Müllerian duct (MD) is the anlage of the oviduct, uterus and upper part of the vagina, the main parts of the female reproductive tract. Several wingless-type mouse mammary tumor virus (MMTV) integration site family member (Wnt) genes, including Wnt4, Wnt5a and Wnt7a, are involved in the development of MD and its derivatives, with Wnt4 particularly critical, since the MD fails to develop in its absence. We use, here, Wnt4(EGFPCre)-based fate mapping to demonstrate that the MD tip cells and the subsequent MD cells are derived from Wnt4+ lineage cells. Moreover, Wnt4 is required for the initiation of MD-forming cell migration. Application of anti-Wnt4 function-blocking antibodies after the initiation of MD elongation indicated that Wnt4 is necessary for the elongation as well, and consistent with this, cell culture wound-healing assays with NIH3T3 cells overexpressing Wnt4 promoted cell migration by comparison with controls. In contrast to the Wnt4 null embryos, some Wnt4(monomeric cherry/monomeric cherry) (Wnt4(mCh/mCh)) hypomorphic mice survived to adulthood and formed MD in ∼45% of cases. Nevertheless, the MD of the Wnt4(mCh/mCh) females had altered cell polarization and basement membrane deposition relative to the controls. Examination of the reproductive tract of the Wnt4(mCh/mCh) females indicated a poorly coiled oviduct, absence of the endometrial glands and an undifferentiated myometrium, and these mice were prone to develop a hydro-uterus. In conclusion, the results suggest that the Wnt4 gene encodes signals that are important for various aspects of female reproductive tract development.
Collapse
Affiliation(s)
- Renata Prunskaite-Hyyryläinen
- Oulu Centre for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland and
| | - Ilya Skovorodkin
- Oulu Centre for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland and
| | - Qi Xu
- Oulu Centre for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland and
| | | | - Jingdong Shan
- Oulu Centre for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland and
| | - Seppo J Vainio
- Oulu Centre for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Laboratory of Developmental Biology, InfoTech Oulu, University of Oulu, PO Box 5000, FIN-90014 Oulu, Finland and
| |
Collapse
|
35
|
Ariza L, Carmona R, Cañete A, Cano E, Muñoz-Chápuli R. Coelomic epithelium-derived cells in visceral morphogenesis. Dev Dyn 2015; 245:307-22. [DOI: 10.1002/dvdy.24373] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Affiliation(s)
- Laura Ariza
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Rita Carmona
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Ana Cañete
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| | - Elena Cano
- Integrative Vascular Biology Lab, Max Delbrück Center for Molecular Medicine; Robert-Rössle-Str. 10 13092, Berlin Germany
| | - Ramón Muñoz-Chápuli
- University of Málaga, Faculty of Science, Department of Animal Biology; Málaga Spain
- Andalusian Center for Nanomedicine and Biotechnology (BIONAND); Campanillas Spain
| |
Collapse
|
36
|
Du J, Hincke MT, Rose-Martel M, Hennequet-Antier C, Brionne A, Cogburn LA, Nys Y, Gautron J. Identifying specific proteins involved in eggshell membrane formation using gene expression analysis and bioinformatics. BMC Genomics 2015; 16:792. [PMID: 26470705 PMCID: PMC4608100 DOI: 10.1186/s12864-015-2013-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/04/2015] [Indexed: 12/16/2022] Open
Abstract
Background The avian eggshell membranes surround the egg white and provide a structural foundation for calcification of the eggshell which is essential for avian reproduction; moreover, it is also a natural biomaterial with many potential industrial and biomedical applications. Due to the insoluble and stable nature of the eggshell membrane fibres, their formation and protein constituents remain poorly characterized. The purpose of this study was to identify genes encoding eggshell membrane proteins, particularly those responsible for its structural features, by analyzing the transcriptome of the white isthmus segment of the oviduct, which is the specialized region responsible for the fabrication of the membrane fibres. Results The Del-Mar 14 K chicken microarray was used to investigate up-regulated expression of transcripts in the white isthmus (WI) compared with the adjacent magnum (Ma) and uterine (Ut) segments of the hen oviduct. Analysis revealed 135 clones hybridizing to over-expressed transcripts (WI/Ma + WI/Ut), and corresponding to 107 NCBI annotated non-redundant Gallus gallus gene IDs. This combined analysis revealed that the structural proteins highly over-expressed in the white isthmus include collagen X (COL10A1), fibrillin-1 (FBN1) and cysteine rich eggshell membrane protein (CREMP). These results validate previous proteomics studies which have identified collagen X (α-1) and CREMP in soluble eggshell extracts. Genes encoding collagen-processing enzymes such as lysyl oxidase homologs 1, 2 and 3 (LOXL1, LOXL2 and LOXL3), prolyl 4 hydroxylase subunit α-2 and beta polypeptide (P4HA2 and P4HB) as well as peptidyl-prolyl cis-trans isomerase C (PPIC) were also over-expressed. Additionally, genes encoding proteins known to regulate disulfide cross-linking, including sulfhydryl oxidase (QSOX1) and thioredoxin (TXN), were identified which suggests that coordinated up-regulation of genes in the white isthmus is associated with eggshell membrane fibre formation. Conclusions The present study has identified genes associated with the processing of collagen, other structural proteins, and disulfide-mediated cross-linking during eggshell membrane formation in the white isthmus. Identification of these genes will provide new insight into eggshell membrane structure and mechanisms of formation that will assist in the development of selection strategies to improve eggshell quality and food safety of the table egg. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2013-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingwen Du
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8 M5, Canada.
| | - Maxwell T Hincke
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8 M5, Canada.
| | - Megan Rose-Martel
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, K1H 8 M5, Canada.
| | | | | | - Larry A Cogburn
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, 19717, USA.
| | - Yves Nys
- INRA, UR83 Recherches Avicoles, F-37380, Nouzilly, France.
| | - Joel Gautron
- INRA, UR83 Recherches Avicoles, F-37380, Nouzilly, France.
| |
Collapse
|
37
|
Georgas KM, Armstrong J, Keast JR, Larkins CE, McHugh KM, Southard-Smith EM, Cohn MJ, Batourina E, Dan H, Schneider K, Buehler DP, Wiese CB, Brennan J, Davies JA, Harding SD, Baldock RA, Little MH, Vezina CM, Mendelsohn C. An illustrated anatomical ontology of the developing mouse lower urogenital tract. Development 2015; 142:1893-908. [PMID: 25968320 DOI: 10.1242/dev.117903] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 04/01/2015] [Indexed: 01/10/2023]
Abstract
Malformation of the urogenital tract represents a considerable paediatric burden, with many defects affecting the lower urinary tract (LUT), genital tubercle and associated structures. Understanding the molecular basis of such defects frequently draws on murine models. However, human anatomical terms do not always superimpose on the mouse, and the lack of accurate and standardised nomenclature is hampering the utility of such animal models. We previously developed an anatomical ontology for the murine urogenital system. Here, we present a comprehensive update of this ontology pertaining to mouse LUT, genital tubercle and associated reproductive structures (E10.5 to adult). Ontology changes were based on recently published insights into the cellular and gross anatomy of these structures, and on new analyses of epithelial cell types present in the pelvic urethra and regions of the bladder. Ontology changes include new structures, tissue layers and cell types within the LUT, external genitalia and lower reproductive structures. Representative illustrations, detailed text descriptions and molecular markers that selectively label muscle, nerves/ganglia and epithelia of the lower urogenital system are also presented. The revised ontology will be an important tool for researchers studying urogenital development/malformation in mouse models and will improve our capacity to appropriately interpret these with respect to the human situation.
Collapse
Affiliation(s)
- Kylie M Georgas
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jane Armstrong
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Janet R Keast
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christine E Larkins
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Kirk M McHugh
- Centre for Molecular and Human Genetics, The Research Institute at Nationwide Children's Hospital and Division of Anatomy, The Ohio State University, Columbus, OH 43205/10, USA
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA Department of Biology, Genetics Institute, University of Florida, Gainesville, FL 32610, USA Howard Hughes Medical Institute, University of Florida, Gainesville, FL 32610, USA
| | | | - Hanbin Dan
- Columbia University, Department of Urology, New York, NY 10032, USA
| | - Kerry Schneider
- Columbia University, Department of Urology, New York, NY 10032, USA
| | - Dennis P Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Carrie B Wiese
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jane Brennan
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Jamie A Davies
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Simon D Harding
- MRC Human Genetics Unit, MRC IGMM, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Richard A Baldock
- MRC Human Genetics Unit, MRC IGMM, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Melissa H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Chad M Vezina
- University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI 53706, USA
| | - Cathy Mendelsohn
- Columbia University, Department of Urology, New York, NY 10032, USA
| |
Collapse
|
38
|
DMRT1 is required for Müllerian duct formation in the chicken embryo. Dev Biol 2015; 400:224-36. [PMID: 25684667 DOI: 10.1016/j.ydbio.2015.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/29/2015] [Accepted: 02/04/2015] [Indexed: 11/22/2022]
Abstract
DMRT1 is a conserved transcription factor with a central role in gonadal sex differentiation. In all vertebrates studied, DMRT1 plays an essential function in testis development and/or maintenance. No studies have reported a role for DMRT1 outside the gonads. Here, we show that DMRT1 is expressed in the paired Müllerian ducts in the chicken embryo, where it is required for duct formation. DMRT1 mRNA and protein are expressed in the early forming Müllerian ridge, and in cells undergoing an epithelial to mesenchyme transition during duct morphogenesis. RNAi-mediated knockdown of DMRT1 in ovo causes a greatly reduced mesenchymal layer, which blocks caudal extension of the duct luminal epithelium. Critical markers of Müllerian duct formation in mammals, Pax2 in the duct epithelium and Wnt4 in the mesenchyme, are conserved in chicken and their expression disrupted in DMRT1 knockdown ducts. We conclude that DMRT1 is required for the early steps of Müllerian duct development. DMRT1 regulates Müllerian ridge and mesenchyme formation and its loss blocks caudal extension of the duct. While DMRT1 plays an important role during testis development and maintenance in many vertebrate species, this is the first report showing a requirement for DMRT1 in Müllerian duct development.
Collapse
|
39
|
Grinspon RP, Rey RA. When hormone defects cannot explain it: malformative disorders of sex development. ACTA ACUST UNITED AC 2014; 102:359-73. [PMID: 25472912 DOI: 10.1002/bdrc.21086] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 10/31/2014] [Indexed: 01/27/2023]
Abstract
The birth of a baby with malformations of the genitalia urges medical action. Even in cases where the condition is not life-threatening, the identification of the external genitalia as male or female is emotionally essential for the family, and genital malformations represent one of the most stressful situations around a newborn. The female or male configuration of the genitalia normally evolves during fetal life according to the genetic, gonadal, and hormonal sex. Disorders of sex development occur when male hormone (androgens and anti-Müllerian hormone) secretion or action is insufficient in the 46,XY fetus or when there is an androgen excess in the 46,XX fetus. However, sex hormone defects during fetal development cannot explain all congenital malformations of the reproductive tract. This review is focused on those congenital conditions in which gonadal function and sex hormone target organ sensitivity are normal and, therefore, not responsible for the genital malformation. Furthermore, because the reproductive and urinary systems share many common pathways in embryo-fetal development, conditions associating urogenital malformations are discussed.
Collapse
Affiliation(s)
- Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos, Aires, Argentina
| | | |
Collapse
|
40
|
Abstract
Uterine anomalies are one of the most common parental causes of recurrent pregnancy loss, occurring in about 19% of patients. Congenital uterine anomalies are most likely caused by HOX gene mutations, although the mechanism is probably polygenic. There are no known environmental causes other than estrogenic endocrine disruptors such as diethylstilbestrol. Acquired uterine anomalies may result from uterine trauma (adhesions) or benign growths of the myometrium (fibroids) or endometrium (polyps). Although randomized controlled trials are lacking, surgical treatment is recommended for repair of uterine septa, and for removal of severe adhesions and submucosal fibroids, especially if no other causes are identified.
Collapse
Affiliation(s)
- Carolyn R Jaslow
- Department of Biology, Rhodes College, 2000 North Parkway, Memphis, TN 38112, USA.
| |
Collapse
|
41
|
Abstract
Congenital anomalies of the upper urinary tract are common and frequently diagnosed on prenatal ultrasound. In the absence of infection, these anomalies are often asymptomatic. This article reviews key features and long-term implications to assist in discussions with families. In contrast, a perinatal renal tumor is rare but extremely alarming. This update on the most common tumors and their treatment is useful in reassuring parents that most infants, after primary surgical resection, are cured without adjuvant therapies. To understand renal agenesis and other congenital renal malformations and their associated anomalies, a brief review of normal renal development is presented.
Collapse
Affiliation(s)
- Ellen Shapiro
- Department of Urology, New York University School of Medicine, 150 32nd Street, 2nd Floor, New York, NY 10017, USA.
| |
Collapse
|
42
|
Mullen RD, Behringer RR. Molecular genetics of Müllerian duct formation, regression and differentiation. Sex Dev 2014; 8:281-96. [PMID: 25033758 DOI: 10.1159/000364935] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Müllerian duct (MD) forms the female reproductive tract (FRT) consisting of the oviducts, uterus, cervix, and upper vagina. FRT function is vital to fertility, providing the site of fertilization, embryo implantation and fetal development. Developmental defects in the formation and diseases of the FRT, including cancer and endometriosis, are prevalent in humans and can result in infertility and death. Furthermore, because the MDs are initially formed regardless of genotypic sex, mesenchymal to epithelial signaling is required in males to mediate MD regression and prevents the development of MD-derived organs. In males, defects in MD regression result in the retention of FRT organs and have been described in several human syndromes. Although to date not reported in humans, ectopic activation of MD regression signaling components in females can result in aplasia of the FRT. Clearly, MD development is important to human health; however, the molecular mechanisms remain largely undetermined. Molecular genetics studies of human diseases and mouse models have provided new insights into molecular signaling during MD development, regression and differentiation. This review will provide an overview of MD development and important genes and signaling mechanisms involved.
Collapse
Affiliation(s)
- Rachel D Mullen
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, Tex., USA
| | | |
Collapse
|
43
|
Dubeau L, Drapkin R. Coming into focus: the nonovarian origins of ovarian cancer. Ann Oncol 2014; 24 Suppl 8:viii28-viii35. [PMID: 24131966 DOI: 10.1093/annonc/mdt308] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The traditional view of epithelial ovarian cancer asserts that all tumor subtypes share a common origin in the ovarian surface epithelium (OSE) DESIGN: A literature review was carried out to summarize the emerging understanding of extraovarian sources of epithelial ovarian carcinomas. RESULTS Historically, there were no diagnostic criteria for documenting the origin of ovarian epithelial carcinomas. Moreover, there are no normal epithelial tissues in the ovary with morphologic similarities to these tumors. In fact, no precursor lesions have ever been reproducibly identified in the ovary. However, there is a strong correlation between extrauterine Müllerian tissue and the development of ovarian carcinomas, tumors of low malignant potential, and cystadenomas. The most recent support for this hypothesis comes from the careful analysis of risk-reducing bilateral salpingo-oopherectomy specimens from BRCA1 or BRCA2 mutation carriers. These studies showed that a significant majority of high-grade serous ovarian carcinomas, the most common subtype, arise from the fallopian tube fimbriae rather than the OSE. CONCLUSIONS Mounting evidence indicates that the vast majority of epithelial ovarian carcinomas are not ovarian in origin. Extrauterine Müllerian epithelium from various sites in the reproductive tract likely accounts for the diverse morphology and behavior of these tumors.
Collapse
Affiliation(s)
- L Dubeau
- Department of Pathology, USC Norris Comprehensive Cancer Center and Hospital, University of Southern California, Los Angeles
| | | |
Collapse
|
44
|
Omotehara T, Smith CA, Mantani Y, Kobayashi Y, Tatsumi A, Nagahara D, Hashimoto R, Hirano T, Umemura Y, Yokoyama T, Kitagawa H, Hoshi N. Spatiotemporal expression patterns of doublesex and mab-3 related transcription factor 1 in the chicken developing gonads and Mullerian ducts. Poult Sci 2014; 93:953-8. [PMID: 24706973 DOI: 10.3382/ps.2013-03672] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Sex of birds is genetically determined by the inheritance of sex chromosomes (ZZ for male and ZW for female), and the Z-linked gene named doublesex and mab-3 related transcription factor 1 (DMRT1) is a candidate sex-determining gene in avian species. However, the mechanisms underlying sex determination in birds are not yet understood, and the expression patterns of the DMRT1 protein in urogenital tissues have not been identified. In the current study, we used immunohistochemistry to investigate the detailed expression patterns of the DMRT1 protein in the urogenital systems (including Müllerian ducts) in male and female chicken embryos throughout embryonic development. Gonadal somatic cells in the male indifferent gonads showed stronger expressions of DMRT1 compared with those in the female indifferent gonads well before the presumptive period of the sex determination, and Sertoli cells forming testicular cords expressed DMRT1 in the testes after sex determination. Germ cells expressed DMRT1 equally in males and females after sex determination. The expression was continuous in males, but in females it gradually disappeared from the germ cells in the central part of the cortex of the left ovary toward both edges. The DMRT1 was also detected in the tubal ridge, which is a precursor of the Müllerian duct, and at the mesenchyme and outermost coelomic epithelium of the Müllerian duct in both sexes. Strong expression was observed in the males, but it was restricted to coelomic epithelium after the regression of the duct started. Thus, we observed the detailed spatiotemporal expression patterns of DMRT1 in the developing chicken urogenital systems throughout embryonic development, suggesting its various roles in the development of urogenital tissues in the chicken embryo.
Collapse
Affiliation(s)
- T Omotehara
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Cutting AD, Ayers K, Davidson N, Oshlack A, Doran T, Sinclair AH, Tizard M, Smith CA. Identification, expression, and regulation of anti-Müllerian hormone type-II receptor in the embryonic chicken gonad. Biol Reprod 2014; 90:106. [PMID: 24621923 DOI: 10.1095/biolreprod.113.116491] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Anti-Müllerian hormone (AMH) signaling is required for proper development of the urogenital system in vertebrates. In male mammals, AMH is responsible for regressing the Müllerian ducts, which otherwise develop into the fallopian tubes, oviducts, and upper vagina of the female reproductive tract. This role is highly conserved across higher vertebrates. However, AMH is required for testis development in fish species that lack Müllerian ducts, implying that AMH signaling has broader roles in other vertebrates. AMH signals through two serine/threonine kinase receptors. The primary AMH receptor, AMH receptor type-II (AMHR2), recruits the type I receptor, which transduces the signal intracellularly. To enhance our understanding of AMH signaling and the potential role of AMH in gonadal sex differentiation, we cloned chicken AMHR2 cDNA and examined its expression profile during gonadal sex differentiation. AMHR2 is expressed in the gonads and Müllerian ducts of both sexes but is more strongly expressed in males after the onset of gonadal sex differentiation. In the testes, the AMHR2 protein colocalizes with AMH, within Sertoli cells of the testis cords. AMHR2 protein expression is up-regulated in female embryos treated with the estrogen synthesis inhibitor fadrozole. Conversely, knockdown of the key testis gene DMRT1 leads to disruption of AMHR2 expression in the developing seminiferous cords of males. These results indicate that AMHR2 is developmentally regulated during testicular differentiation in the chicken embryo. AMH signaling may be important for gonadal differentiation in addition to Müllerian duct regression in birds.
Collapse
Affiliation(s)
- Andrew D Cutting
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Katie Ayers
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Nadia Davidson
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Alicia Oshlack
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Tim Doran
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Andrew H Sinclair
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Mark Tizard
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Craig A Smith
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Department of Zoology, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| |
Collapse
|
46
|
Huang CC, Orvis GD, Kwan KM, Behringer RR. Lhx1 is required in Müllerian duct epithelium for uterine development. Dev Biol 2014; 389:124-36. [PMID: 24560999 DOI: 10.1016/j.ydbio.2014.01.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 02/08/2023]
Abstract
The female reproductive tract organs of mammals, including the oviducts, uterus, cervix and upper vagina, are derived from the Müllerian ducts, a pair of epithelial tubes that form within the mesonephroi. The Müllerian ducts form in a rostral to caudal manner, guided by and dependent on the Wolffian ducts that have already formed. Experimental embryological studies indicate that caudal elongation of the Müllerian duct towards the urogenital sinus occurs in part by proliferation at the ductal tip. The molecular mechanisms that regulate the elongation of the Müllerian duct are currently unclear. Lhx1 encodes a LIM-homeodomain transcription factor that is essential for male and female reproductive tract development. Lhx1 is expressed in both the Wolffian and Müllerian ducts. Wolffian duct-specific knockout of Lhx1 results in degeneration of the Wolffian duct and consequently the non-cell-autonomous loss of the Müllerian duct. To determine the role of Lhx1 specifically in the Müllerian duct epithelium, we performed a Müllerian duct-specific knockout study using Wnt7a-Cre mice. Loss of Lhx1 in the Müllerian duct epithelium led to a block in Müllerian duct elongation and uterine hypoplasia characterized by loss of the entire endometrium (luminal and glandular epithelium and stroma) and inner circular but not the outer longitudinal muscle layer. Time-lapse imaging and molecular analyses indicate that Lhx1 acts cell autonomously to maintain ductal progenitor cells for Müllerian duct elongation. These studies identify LHX1 as the first transcription factor that is essential in the Müllerian duct epithelial progenitor cells for female reproductive tract development. Furthermore, these genetic studies demonstrate the requirement of epithelial-mesenchymal interactions for uterine tissue compartment differentiation.
Collapse
Affiliation(s)
- Cheng-Chiu Huang
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Grant D Orvis
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Kin Ming Kwan
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, PR China
| | - Richard R Behringer
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
47
|
Park JH, Tanaka Y, Arango NA, Zhang L, Benedict LA, Roh MI, Donahoe PK, Teixeira JM. Induction of WNT inhibitory factor 1 expression by Müllerian inhibiting substance/antiMullerian hormone in the Müllerian duct mesenchyme is linked to Müllerian duct regression. Dev Biol 2013; 386:227-36. [PMID: 24362065 DOI: 10.1016/j.ydbio.2013.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 01/07/2023]
Abstract
A key event during mammalian sexual development is regression of the Müllerian ducts (MDs) in the bipotential urogenital ridges (UGRs) of fetal males, which is caused by the expression of Müllerian inhibiting substance (MIS) in the Sertoli cells of the differentiating testes. The paracrine signaling mechanisms involved in MD regression are not completely understood, particularly since the receptor for MIS, MISR2, is expressed in the mesenchyme surrounding the MD, but regression occurs in both the epithelium and mesenchyme. Microarray analysis comparing MIS signaling competent and Misr2 knockout embryonic UGRs was performed to identify secreted factors that might be important for MIS-mediated regression of the MD. A seven-fold increase in the expression of Wif1, an inhibitor of WNT/β-catenin signaling, was observed in the Misr2-expressing UGRs. Whole mount in situ hybridization of Wif1 revealed a spatial and temporal pattern of expression consistent with Misr2 during the window of MD regression in the mesenchyme surrounding the MD epithelium that was absent in both female UGRs and UGRs knocked out for Misr2. Knockdown of Wif1 expression in male UGRs by Wif1-specific siRNAs beginning on embryonic day 13.5 resulted in MD retention in an organ culture assay, and exposure of female UGRs to added recombinant human MIS induced Wif1 expression in the MD mesenchyme. Knockdown of Wif1 led to increased expression of β-catenin and its downstream targets TCF1/LEF1 in the MD mesenchyme and to decreased apoptosis, resulting in partial to complete retention of the MD. These results strongly suggest that WIF1 secretion by the MD mesenchyme plays a role in MD regression in fetal males.
Collapse
Affiliation(s)
- Joo Hyun Park
- Vincent Center of Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Yoshihiro Tanaka
- Vincent Center of Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Nelson A Arango
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Lihua Zhang
- Vincent Center of Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| | - L Andrew Benedict
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Mi In Roh
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Jose M Teixeira
- Vincent Center of Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
48
|
Osman AHK, Farouk SM, Eidaroos H, Abbott LC, Ahmed AA. Prenatal metanephrogenesis of the camel: morphological evidence of epithelial-mesenchymal interaction. Anat Histol Embryol 2013; 43:141-52. [PMID: 23713903 DOI: 10.1111/ahe.12057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/03/2013] [Indexed: 11/30/2022]
Abstract
The present investigation examined histogenesis of epithelial, stromal and angiogenic elements of the prenatal camel permanent or metanephric kidney. The primitive metanephros was first observed at the 13-mm crown vertebral rump length (CVRL) stage as an ovoid structure composed of a centrally located epithelial ureteric bud and peripheral circumscribed masses of undifferentiated mesenchymal cells. The first morphological evidence of glomerulogenesis was observed at the 28-mm CVRL stage. Developing renal corpuscles became obvious at the 35-mm CVRL stage. At the 60-mm CVRL stage, the epithelial renal pelvis gave rise to tubular branches that extended towards the cortical zone. These branches represented the presumptive collecting ducts. Differentiation of renal tubules into the proximal and distal convoluted tubules was observed at the 95-mm CVRL stage. At the 130-mm CVRL stage, the renal medulla was clearly delineated into medullary pyramids, which in association with the corresponding cortical caps formed the morphological basis of the renal lobar formation. A gradual nephrogenic decline was noticed from the 940-mm CVRL on; however, the process of nephrogenesis persisted throughout all the studied foetal stages.
Collapse
Affiliation(s)
- A-H K Osman
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41511, Egypt
| | | | | | | | | |
Collapse
|
49
|
Paik DY, Janzen DM, Schafenacker AM, Velasco VS, Shung MS, Cheng D, Huang J, Witte ON, Memarzadeh S. Stem-like epithelial cells are concentrated in the distal end of the fallopian tube: a site for injury and serous cancer initiation. Stem Cells 2013; 30:2487-97. [PMID: 22911892 PMCID: PMC4442093 DOI: 10.1002/stem.1207] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The reproductive role of the fallopian tube is to transport the sperm and egg. The tube is positioned to act as a bridge between the ovary where the egg is released and the uterus where implantation occurs. Throughout reproductive years, the fallopian tube epithelium undergoes repetitive damage and regeneration. Although a reservoir of adult epithelial stem cells must exist to replenish damaged cells, they remain unidentified. Here, we report isolation of a subset of basally located human fallopian tube epithelia (FTE) that lack markers of ciliated (β-tubulin; TUBB4) or secretory (PAX8) differentiated cells. These undifferentiated cells expressed cell surface antigens: epithelial cell adhesion molecule, CD44, and integrin α 6. This FTE subpopulation was fivefold enriched for cells capable of clonal growth and self-renewal suggesting that they contain the FTE stem-like cells (FTESCs). A twofold enrichment of the FTESC was found in the distal compared to the proximal end of the tube. The distal fimbriated end of the fallopian tube is a well-characterized locus for initiation of serous carcinomas. An expansion of the cells expressing markers of FTESC was detected in tubal intraepithelial carcinomas and in fallopian tubes from patients with invasive serous cancer. These findings suggest that FTESC may play a role in the initiation of serous tumors. Characterization of these stem-like cells will provide new insight into how the FTE regenerate, respond to injury, and may initiate cancer.
Collapse
Affiliation(s)
- Daniel Y Paik
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Patterson AL, Zhang L, Arango NA, Teixeira J, Pru JK. Mesenchymal-to-epithelial transition contributes to endometrial regeneration following natural and artificial decidualization. Stem Cells Dev 2013; 22:964-74. [PMID: 23216285 DOI: 10.1089/scd.2012.0435] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Despite being a histologically dynamic organ, mechanisms coordinating uterine regeneration during the menstrual/estrous cycle and following parturition are poorly understood. In the current study, we hypothesized that endometrial epithelial tissue regeneration is accomplished, in part, by mesenchymal-to-epithelial transition (MET). To test this hypothesis, fate mapping studies were completed using a double transgenic (Tg) reporter strain, Amhr2-Cre; Rosa26-Stop(fl/fl-EYFP) (i.e., flox-stop EYFP reporter). EYFP expression was observed in Müllerian duct mesenchyme-derived stroma and myometrium, but not epithelia in young and peripubertal double Tg female mice. However, mosaic EYFP expression was observed in epithelia of double Tg mice after parturition. To ensure the observed epithelial EYFP expression was not due to leaky Amhr2 promoter activity, resulting in aberrant Cre expression, transgenic mice expressing LacZ under the control of the Amhr2 promoter (Amhr2-LacZ) were used to monitor β-galactosidase (β-Gal) activity within the uterus. β-Gal activity was not detected in luminal or glandular epithelia regardless of age, reproductive status, or degree of damage incurred within the uterus. Lastly, a unique population of transitional cells was identified that expressed the epithelial cell marker, pan-cytokeratin, and the stromal cell marker, vimentin. These cells localized predominantly to the regeneration zone in the mesometrial region of the endometrium. These findings suggest a previously unappreciated role for MET in endometrial regeneration and have important implications for proliferative diseases of the endometrium such as endometriosis.
Collapse
Affiliation(s)
- Amanda L Patterson
- Department of Animal Science, Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164, USA
| | | | | | | | | |
Collapse
|