1
|
Liu Z, Luo X, Zhang Z, Zhang Q, Wang C, Chen H, Long C, Liu X, Wei G. MAFB-mediated CEBPA regulated human urothelium growth through Wnt/β-catenin signaling pathway. Genes Dis 2025; 12:101432. [PMID: 39569391 PMCID: PMC11577151 DOI: 10.1016/j.gendis.2024.101432] [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: 03/20/2024] [Accepted: 08/29/2024] [Indexed: 11/22/2024] Open
Abstract
MAFB is essential for regulating male-type urethral differentiation, and especially, its variation can contribute to hypospadias in mice. However, the potential mechanism is still unclear. Here we observed that the basic leucine zipper (bZIP) transcription factor MAFB and CCAAT/enhancer-binding protein alpha (CEBPA) could promote human urothelium SV-HUC-1 growth. Moreover, MAFB and CEBPA expression were reduced in the prepuce tissues of hypospadias patients. Based on transcriptome sequencing analysis and Western blot, MAFB knockdown was found to suppress CEBPA protein expression and repress Wnt/β-catenin signaling in urothelium cells. Meanwhile, we observed blocked cell-cycle progression from the G1 to the S phase, inhibited cell proliferation, and activated apoptosis. Furthermore, MAFB could facilitate CEBPA transcription and regulate the proliferation of urothelium. The above results indicated that MAFB-mediated inhibition of urothelial SV-HUC-1 growth resulted from inhibiting the Wnt/β-catenin signaling pathway by down-regulating CEBPA. Our findings provide new insight into the understanding of genes associated with hypospadias and the pathogenic mechanism of this disorder.
Collapse
Affiliation(s)
- Zhenmin Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Xingguo Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Zhicheng Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Qiang Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Chong Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Hongsong Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Chunlan Long
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Xing Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| |
Collapse
|
2
|
Formstone C, Aldeiri B, Davenport M, Francis-West P. Ventral body wall closure: Mechanistic insights from mouse models and translation to human pathology. Dev Dyn 2024. [PMID: 39319771 DOI: 10.1002/dvdy.735] [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: 12/19/2023] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
The ventral body wall (VBW) that encloses the thoracic and abdominal cavities arises by extensive cell movements and morphogenetic changes during embryonic development. These morphogenetic processes include embryonic folding generating the primary body wall; the initial ventral cover of the embryo, followed by directed mesodermal cell migrations, contributing to the secondary body wall. Clinical anomalies in VBW development affect approximately 1 in 3000 live births. However, the cell interactions and critical cellular behaviors that control VBW development remain little understood. Here, we describe the embryonic origins of the VBW, the cellular and morphogenetic processes, and key genes, that are essential for VBW development. We also provide a clinical overview of VBW anomalies, together with environmental and genetic influences, and discuss the insight gained from over 70 mouse models that exhibit VBW defects, and their relevance, with respect to human pathology. In doing so we propose a phenotypic framework for researchers in the field which takes into account the clinical picture. We also highlight cases where there is a current paucity of mouse models for particular clinical defects and key gaps in knowledge about embryonic VBW development that need to be addressed to further understand mechanisms of human VBW pathologies.
Collapse
Affiliation(s)
- Caroline Formstone
- Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, Hatfield, UK
| | - Bashar Aldeiri
- Department of Paediatric Surgery, Chelsea and Westminster Hospital, London, UK
| | - Mark Davenport
- Department of Paediatric Surgery, King's College Hospital, London, UK
| | | |
Collapse
|
3
|
Lozovska A, Korovesi AG, Dias A, Lopes A, Fowler DA, Martins GG, Nóvoa A, Mallo M. Tgfbr1 controls developmental plasticity between the hindlimb and external genitalia by remodeling their regulatory landscape. Nat Commun 2024; 15:2509. [PMID: 38509075 PMCID: PMC10954616 DOI: 10.1038/s41467-024-46870-z] [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: 06/09/2023] [Accepted: 03/13/2024] [Indexed: 03/22/2024] Open
Abstract
The hindlimb and external genitalia of present-day tetrapods are thought to derive from an ancestral common primordium that evolved to generate a wide diversity of structures adapted for efficient locomotion and mating in the ecological niche occupied by the species. We show that despite long evolutionary distance from the ancestral condition, the early primordium of the mouse external genitalia preserved the capacity to take hindlimb fates. In the absence of Tgfbr1, the pericloacal mesoderm generates an extra pair of hindlimbs at the expense of the external genitalia. It has been shown that the hindlimb and the genital primordia share many of their key regulatory factors. Tgfbr1 controls the response to those factors by modulating the accessibility status of regulatory elements that control the gene regulatory networks leading to the formation of genital or hindlimb structures. Our work uncovers a remarkable tissue plasticity with potential implications in the evolution of the hindlimb/genital area of tetrapods, and identifies an additional mechanism for Tgfbr1 activity that might also contribute to the control of other physiological or pathological processes.
Collapse
Affiliation(s)
- Anastasiia Lozovska
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Artemis G Korovesi
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - André Dias
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Alexandre Lopes
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Donald A Fowler
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Gabriel G Martins
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Ana Nóvoa
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Moisés Mallo
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal.
| |
Collapse
|
4
|
Haller M, Yin Y, Haller G, Li T, Li Q, Lamb LE, Ma L. Streamlined identification of clinically and functionally relevant genetic regulators of lower-tract urogenital development. Proc Natl Acad Sci U S A 2024; 121:e2309466121. [PMID: 38300866 PMCID: PMC10861909 DOI: 10.1073/pnas.2309466121] [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: 06/06/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024] Open
Abstract
Congenital anomalies of the lower genitourinary (LGU) tract are frequently comorbid due to genetically linked developmental pathways, and are among the most common yet most socially stigmatized congenital phenotypes. Genes involved in sexual differentiation are prime candidates for developmental anomalies of multiple LGU organs, but insufficient prospective screening tools have prevented the rapid identification of causative genes. Androgen signaling is among the most influential modulators of LGU development. The present study uses SpDamID technology in vivo to generate a comprehensive map of the pathways actively regulated by the androgen receptor (AR) in the genitalia in the presence of the p300 coactivator, identifying wingless/integrated (WNT) signaling as a highly enriched AR-regulated pathway in the genitalia. Transcription factor (TF) hits were then assayed for sexually dimorphic expression at two critical time points and also cross-referenced to a database of clinically relevant copy number variations to identify 252 TFs exhibiting copy variation in patients with LGU phenotypes. A subset of 54 TFs was identified for which LGU phenotypes are statistically overrepresented as a proportion of total observed phenotypes. The 252 TF hitlist was then subjected to a functional screen to identify hits whose silencing affects genital mesenchymal growth rates. Overlap of these datasets results in a refined list of 133 TFs of both functional and clinical relevance to LGU development, 31 of which are top priority candidates, including the well-documented renal progenitor regulator, Sall1. Loss of Sall1 was examined in vivo and confirmed to be a powerful regulator of LGU development.
Collapse
Affiliation(s)
- Meade Haller
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Yan Yin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Gabe Haller
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO63110
| | - Tian Li
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Qiufang Li
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Laura E. Lamb
- Department of Urology, William Beaumont School of Medicine, Oakland University, Rochester, MI48309
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
5
|
Hashimoto D, Fujimoto K, Nakata M, Suzuki T, Kumegawa S, Ueda Y, Suzuki K, Asamura S, Yamada G. Developmental and functional roles of androgen and interactive signals for external genitalia and erectile tissues. Reprod Med Biol 2024; 23:e12611. [PMID: 39372370 PMCID: PMC11456227 DOI: 10.1002/rmb2.12611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Background Recent progress in molecular and signal analyses revealed essential functions of cellular signals including androgen and related growth factors such as Wnt regulators for external genitalia (ExG) development and its pathogenesis. Accumulated data showed their fundamental functions also for erectile tissue (corporal body) development and its abnormalities. The current review focuses on such signals from developmental and functional viewpoints. Methods Experimental strategies including histological and molecular signal analyses with conditional mutant mice for androgen and Wnt signals have been extensively utilized. Main findings Essential roles of androgen for the development of male-type ExG and urethral formation are shown. Wnt signals are associated with androgen for male-type ExG organogenesis. Androgen plays essential roles in the development of erectile tissue, the corporal body and it also regulates the duration time of erection. Wnt and other signals are essential for the regulation of mesenchymal cells of erectile tissue as shown by its conditional mutant mouse analyses. Stress signals, continuous erection, and the potential of lymphatic characteristics of the erectile vessels with sinusoids are also shown. Conclusion Reiterated involvement of androgen, Wnt, and other regulatory factors is stated for the development and pathogenesis of ExG and erectile tissues.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of UrologyUrological Science Institute, Yonsei University College of MedicineSeoulSouth Korea
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Masanori Nakata
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuya Suzuki
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shinji Kumegawa
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Yuko Ueda
- Department of UrologyOsaka Women's and Children's HospitalOsakaJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Gen Yamada
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
6
|
Tan H, Zheng Z, Wang S, Yang L, Widelka M, Chen D. Neonatal exposure to bisphenol analogues disrupts genital development in male mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 330:121783. [PMID: 37164221 DOI: 10.1016/j.envpol.2023.121783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023]
Abstract
The public concern and governmental regulations on bisphenol A (BPA) have stimulated the development and production of alternative analogues to replace BPA in a myriad of applications. Given the endocrine disrupting activities of BPA and potentially other analogues, the present study investigated and compared the effects of neonatal exposure to BPA, BPB, BPE, BPF, and BPS on the genital development in male mice. Pups were injected subcutaneously on the right shoulder in the mornings of postnatal days P0.5, P2, P4, and P6, resulting in a low dose of 0.05 μg/g body weight (bw)/day and a high dose of 10 μg/g bw/day. Mice were sacrificed at predetermined time and evaluated for gene expression levels (3 days after birth or P3), steroid hormone levels (P5), and morphological changes (P21). The results demonstrated that BPA, BPB, BPE, or BPF significantly shortened glans penis length and anogenital distance, while BPS didn't. Testis weight and anogenital distance were also significantly affected by BPA, BPE or BPF. The results also revealed that bisphenol analogues exposure significantly reduced testosterone levels, and altered the expression levels of developmental genes networks in developing penis of mice. Our data demonstrate that selected bisphenol analogues may possess similar endocrine disrupting effects compared to BPA, and exposure to these analogues could affect reproductive development of male mice. This raises the concern on the environmental and health safety of bisphenol analogues applied as industrial BPA replacements.
Collapse
Affiliation(s)
- Hongli Tan
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China
| | - Zhengui Zheng
- Department of Physiology, Southern Illinois University, School of Medicine, Carbondale, IL, 62901, United States
| | - Shanshan Wang
- Department of Physiology, Southern Illinois University, School of Medicine, Carbondale, IL, 62901, United States
| | - Liu Yang
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China
| | - Margaret Widelka
- Department of Physiology, Southern Illinois University, School of Medicine, Carbondale, IL, 62901, United States
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
7
|
Furuminato K, Minatoya S, Senoo E, Goto T, Yamazaki S, Sakaguchi M, Toyota K, Iguchi T, Miyagawa S. The role of mesenchymal estrogen receptor 1 in mouse uterus in response to estrogen. Sci Rep 2023; 13:12293. [PMID: 37516793 PMCID: PMC10387046 DOI: 10.1038/s41598-023-39474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023] Open
Abstract
Estrogens play important roles in uterine growth and homeostasis through estrogen receptors (ESR1 and ESR2). To address the role of ESR1-mediated tissue events in the murine uterus, we analyzed mice with a mesenchymal tissue-specific knockout of Esr1. Isl1-driven Cre expression generated Esr1 deletion in the uterine stroma and endometrium (Isl-Esr1KO). We showed that overall structure of the Isl1-Esr1KO mouse uterus developed normally, but estrogen responsiveness and subsequent growth were defective, suggesting that mesenchymal ESR1 is necessary for both epithelial and mesenchymal cell proliferation. Furthermore, RNA-seq analysis revealed that the majority of estrogen-induced genes were regulated by stromal ESR1. In control mice, E2 administration induced 9476 up-regulated differentially expressed genes (DEGs), whereas only 1801 up-regulated DEGs were induced by E2 in Isl1-Esr1KO mice. We further showed that stromal ESR1-regulated genes in the mouse uterus included several growth factors and cytokines, which are potential factors that regulate epithelial and stromal tissue interaction, and also genes involved in lipid homeostasis. Therefore, we infer that stromal ESR1 expression is indispensable for most estrogen actions in the mouse uterus and the current results provide new insights into estrogen-mediated homeostasis in female reproductive organs.
Collapse
Affiliation(s)
- Keita Furuminato
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Saki Minatoya
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Eriko Senoo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Tatsuki Goto
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Sho Yamazaki
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Moeka Sakaguchi
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
| | - Kenji Toyota
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto, Ishikawa, 927-0552, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa, 236-0027, Japan
| | - Shinichi Miyagawa
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku 6-3-1, Katsushika, Tokyo, 125-8585, Japan.
| |
Collapse
|
8
|
Tanaka K, Matsumaru D, Suzuki K, Yamada G, Miyagawa S. The role of p63 in embryonic external genitalia outgrowth in mice. Dev Growth Differ 2023; 65:132-140. [PMID: 36680528 PMCID: PMC11520970 DOI: 10.1111/dgd.12840] [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: 08/28/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
Embryonic external genitalia (genital tubercle [GT]) protrude from the cloaca and outgrow as cloacal development progresses. Individual gene functions and knockout phenotypes in GT development have been extensively analyzed; however, the interactions between these genes are not fully understood. In this study, we investigated the role of p63, focusing on its interaction with the Shh-Wnt/Ctnnb1-Fgf8 pathway, a signaling network that is known to play a role in GT outgrowth. p63 was expressed in the epithelial tissues of the GT at E11.5, and the distal tip of the GT predominantly expressed the ΔNp63α isoform. The GTs in p63 knockout embryos had normal Shh expression, but CTNNB1 protein and Fgf8 gene expression in the distal urethral epithelium was decreased or lost. Constitutive expression of CTNNB1 in p63-null embryos restored Fgf8 expression, accompanied by small bud structure development; however, such bud structures could not be maintained by E13.5, at which point mutant GTs exhibited severe abnormalities showing a split shape with a hemorrhagic cloaca. Therefore, p63 is a key component of the signaling pathway that triggers Fgf8 expression in the distal urethral epithelium and contributes to GT outgrowth by ensuring the structural integrity of the cloacal epithelia. Altogether, we propose that p63 plays an essential role in the signaling network for the development of external genitalia.
Collapse
Affiliation(s)
- Kosei Tanaka
- Department of Biological Science and Technology, Faculty of Advances EngineeringTokyo University of ScienceKatsushikaJapan
| | - Daisuke Matsumaru
- Laboratory of Hygienic Chemistry and Molecular ToxicologyGifu Pharmaceutical UniversityGifuJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Gen Yamada
- Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
| | - Shinichi Miyagawa
- Department of Biological Science and Technology, Faculty of Advances EngineeringTokyo University of ScienceKatsushikaJapan
- Division of Biological Environment Innovation, Research Institute for Science and TechnologyTokyo University of ScienceKatsushikaJapan
| |
Collapse
|
9
|
Yin Y, Haller M, Li T, Ma L. Development of an in-vitro high-throughput screening system to identify modulators of genitalia development. PNAS NEXUS 2023; 2:pgac300. [PMID: 36712925 PMCID: PMC9832959 DOI: 10.1093/pnasnexus/pgac300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Sexually dimorphic outgrowth and differentiation of the embryonic genital tubercles (GTs) give rise to the penis in males and the clitoris in females. Defects in androgen production or in response to androgen signaling can lead to various congenital penile anomalies in both mice and humans. Due to lack of a high-throughput screening system, identification of crucial regulators of GT sexual differentiation has been slow. To overcome this research barrier, we isolated embryonic GT mesenchymal (GTme) cells to model genitalia growth and differentiation in vitro. Using either a mechanical or fluorescence-activated cell sorting-assisted purification method, GTme cells were isolated and assayed for their proliferation using a microscopy and image analysis system, on a single cell level over time. Male and female GTme cells inherently exhibit different cellular dynamics, consistent with their in-vivo behaviors. This system allows for the rapid quantitative analyses of numerous drug treatments, and enables the discovery of potential genetic modulators of GT morphogenesis on a large scale. Using this system, we completed a 438-compound library screen and identified 82 kinase inhibitor hits. In mice, in-utero exposure to one such candidate kinase inhibitor, Cediranib, resulted in embryos with severe genitalia defects, especially in males. Gene silencing by RNAi was optimized in this system, laying the foundation for future larger-scale genetic screenings. These findings demonstrate the power of this novel high-throughput system to rapidly and successfully identify modulators of genitalia growth and differentiation, expanding the toolbox for the study of functional genomics and environmental factors.
Collapse
Affiliation(s)
- Yan Yin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Meade Haller
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Tian Li
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Ipulan-Colet LA. Sexual dimorphism through androgen signaling; from external genitalia to muscles. Front Endocrinol (Lausanne) 2022; 13:940229. [PMID: 35983512 PMCID: PMC9379613 DOI: 10.3389/fendo.2022.940229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Sexual dimorphisms can be seen in many organisms with some exhibiting subtle differences while some can be very evident. The difference between male and female can be seen on the morphological level such as discrepancies in body mass, presence of body hair in distinct places, or through the presence of specific reproductive structures. It is known that the development of the reproductive structures is governed by hormone signaling, most commonly explained through the actions of androgen signaling. The developmental program of the male and female external genitalia involves a common anlage, the genital tubercle or GT, that later on develop into a penis and clitoris, respectively. Androgen signaling involvement can be seen in the different tissues in the GT that express Androgen receptor and the different genes that are regulated by androgen in the mesenchyme and endoderm component of the GT. Muscles are also known to be responsive to androgen signaling with male and female muscles exhibiting different capabilities. However, the occurrence of sexual dimorphism in muscle development is unclear. In this minireview, a summary on the role of androgen in the sexually dimorphic development of the genital tubercle was provided. This was used as a framework on analyzing the different mechanism employed by androgen signaling to regulate the sexual dimorphism in muscle development.
Collapse
|
11
|
Tarulli GA, Cripps SM, Pask AJ, Renfree MB. Spatiotemporal map of key signaling factors during early penis development. Dev Dyn 2021; 251:609-624. [PMID: 34697862 PMCID: PMC9539974 DOI: 10.1002/dvdy.433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/27/2021] [Accepted: 09/28/2021] [Indexed: 12/31/2022] Open
Abstract
The formation of the external genitalia is a highly complex developmental process, considering it involves a wide range of cell types and results in sexually dimorphic outcomes. Development is controlled by several secreted signalling factors produced in complex spatiotemporal patterns, including the hedgehog (HH), bone morphogenic protein (BMP), fibroblast growth factor (FGF) and WNT signalling families. Many of these factors act on or are influenced by the actions of the androgen receptor (AR) that is critical to masculinisation. This complexity of expression makes it difficult to conceptualise patterns of potential importance. Mapping expression during key stages of development is needed to develop a comprehensive model of how different cell types interact in formation of external genitalia, and the global regulatory networks at play. This is particularly true in light of the sensitivity of this process to environmental disruption during key stages of development. The goal of this review is to integrate all recent studies on gene expression in early penis development to create a comprehensive spatiotemporal map. This serves as a resource to aid in visualising potentially significant interactions involved in external genital development. Diagrams of published RNA and protein localisation data for key secreted signalling factors during early penis development. Unconventional expression patterns are identified that suggest novel signalling axes during development. Key research gaps and limitations are identified and discussed.
Collapse
Affiliation(s)
- Gerard A Tarulli
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Samuel M Cripps
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Gredler ML, Patterson SE, Seifert AW, Cohn MJ. Foxa1 and Foxa2 orchestrate development of the urethral tube and division of the embryonic cloaca through an autoregulatory loop with Shh. Dev Biol 2020; 465:23-30. [PMID: 32645357 DOI: 10.1016/j.ydbio.2020.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/04/2023]
Abstract
Congenital anomalies of external genitalia affect approximately 1 in 125 live male births. Development of the genital tubercle, the precursor of the penis and clitoris, is regulated by the urethral plate epithelium, an endodermal signaling center. Signaling activity of the urethral plate is mediated by Sonic hedgehog (SHH), which coordinates outgrowth and patterning of the genital tubercle by controlling cell cycle kinetics and expression of downstream genes. The mechanisms that govern Shh transcription in urethral plate cells are largely unknown. Here we show that deletion of Foxa1 and Foxa2 results in persistent cloaca, an incomplete separation of urinary, genital, and anorectal tracts, and severe hypospadias, a failure of urethral tubulogenesis. Loss of Foxa2 and only one copy of Foxa1 results in urethral fistula, an additional opening of the penile urethra. Foxa1/a2 participate in an autoregulatory feedback loop with Shh, in which FOXA1 and FOXA2 positively regulate transcription of Shh in the urethra, and SHH feeds back to negatively regulate Foxa1 and Foxa2 expression. These findings reveal novel roles for Foxa genes in development of the urethral tube and in division of the embryonic cloaca.
Collapse
Affiliation(s)
- Marissa L Gredler
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA; Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA
| | - Sara E Patterson
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA
| | - Ashley W Seifert
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA
| | - Martin J Cohn
- Department of Biology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA; Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, PO Box 103610, Gainesville, FL, 32611, USA.
| |
Collapse
|
13
|
Johansson HK, Svingen T. Hedgehog signal disruption, gonadal dysgenesis and reproductive disorders: Is there a link to endocrine disrupting chemicals? Curr Res Toxicol 2020; 1:116-123. [PMID: 34345840 PMCID: PMC8320607 DOI: 10.1016/j.crtox.2020.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 01/04/2023] Open
Abstract
Developmental exposure to chemicals that can disrupt sex hormone signaling may cause a broad spectrum of reproductive disorders. This is because reproductive development is tightly regulated by steroid sex hormones. Consequently, non-animal screening methods currently used to test chemicals for potential endocrine disrupting activities typically include steroidogenesis and nuclear receptor assays. In many cases there is a correlation between in vitro and in vivo data examining endocrine disruption, for example between blocked androgen receptor activity and feminized male genitals. However, there are many examples where there is poor, or no, correlation between in vitro data and in vivo effect outcomes in rodent studies, for various reasons. One possible, and less studied, reason for discordance between in vitro and in vivo data is that the mechanisms causing the in vivo effects are not covered by those typically tested for in vitro. This knowledge gap must be addressed if we are to elaborate robust testing strategies that do not rely on animal experimentation. In this review, we highlight the Hedgehog (HH) signaling pathway as a target for environmental chemicals and its potential implications for reproductive disorders originating from early life exposure. A central proposition is that, by disrupting HH signal transduction during critical stages of mammalian development, the endocrine cells of the testes or ovaries fail to develop normally, which ultimately will lead to disrupted sex hormone synthesis and sexual development in both sexes. If this is the case, then such mechanism must also be included in future test strategies aimed at eliminating chemicals that may cause reproductive disorders in humans.
Collapse
Affiliation(s)
- Hanna K.L. Johansson
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| |
Collapse
|
14
|
Wang S, Lawless J, Zheng Z. Prenatal low-dose methyltestosterone, but not dihydrotestosterone, treatment induces penile formation in female mice and guinea pigs†. Biol Reprod 2020; 102:1248-1260. [PMID: 32219310 PMCID: PMC7253790 DOI: 10.1093/biolre/ioaa035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/04/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
Genital tubercle has bisexual potential before sex differentiation. Females exposed to androgen during sex differentiation show masculinized external genitalia, but the effects of different androgens on tubular urethral and penile formation in females are mostly unknown. In this study, we compared the masculinization effects of commonly used androgens methyltestosterone, dihydrotestosterone, and testosterone on the induction of penile formation in females. Our results suggested that prenatal treatment with low doses of methyltestosterone, but not same doses of dihydrotestosterone or testosterone, could induce penile formation in female mice. The minimum dose of dihydrotestosterone and testosterone for inducing tubular urethral formation in female mice was, respectively, 50 and 20 times higher than that of methyltestosterone. In vivo methyltestosterone treatment induced more nuclear translocation of androgen receptors in genital tubercles of female mice, affected Wnt signaling gene expressions, and then led to similar patterns of cell proliferation and death in developing genital tubercles to those of control males. We further revealed that low-dose methyltestosterone, but not same dose of dihydrotestosterone or testosterone, treatment induced penile formation in female guinea pigs. Exposure of female mouse genital tubercle organ culture to methyltestosterone, dihydrotestosterone, or testosterone could induce nuclear translocation of androgen receptors, suggesting that the differential effect of the three androgens in vivo might be due to the hormonal profile in mother or fetus, rather than the local genital tissue. To understand the differential role of these androgens in masculinization process involved is fundamental to androgen replacement therapy for diseases related to external genital masculinization.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL, USA
| | - John Lawless
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL, USA
| | - Zhengui Zheng
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL, USA
| |
Collapse
|
15
|
Chen Y, Renfree MB. Hormonal and Molecular Regulation of Phallus Differentiation in a Marsupial Tammar Wallaby. Genes (Basel) 2020; 11:genes11010106. [PMID: 31963388 PMCID: PMC7017150 DOI: 10.3390/genes11010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/24/2019] [Accepted: 01/14/2020] [Indexed: 11/16/2022] Open
Abstract
Congenital anomalies in phalluses caused by endocrine disruptors have gained a great deal of attention due to its annual increasing rate in males. However, the endocrine-driven molecular regulatory mechanism of abnormal phallus development is complex and remains largely unknown. Here, we review the direct effect of androgen and oestrogen on molecular regulation in phalluses using the marsupial tammar wallaby, whose phallus differentiation occurs after birth. We summarize and discuss the molecular mechanisms underlying phallus differentiation mediated by sonic hedgehog (SHH) at day 50 pp and phallus elongation mediated by insulin-like growth factor 1 (IGF1) and insulin-like growth factor binding protein 3 (IGFBP3), as well as multiple phallus-regulating genes expressed after day 50 pp. We also identify hormone-responsive long non-coding RNAs (lncRNAs) that are co-expressed with their neighboring coding genes. We show that the activation of SHH and IGF1, mediated by balanced androgen receptor (AR) and estrogen receptor 1 (ESR1) signalling, initiates a complex regulatory network in males to constrain the timing of phallus differentiation and to activate the downstream genes that maintain urethral closure and phallus elongation at later stages.
Collapse
Affiliation(s)
- Yu Chen
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32603, USA
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence: (Y.C.); (M.B.R.)
| | - Marilyn B. Renfree
- School of BioSciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence: (Y.C.); (M.B.R.)
| |
Collapse
|
16
|
Kajioka D, Suzuki K, Nakada S, Matsushita S, Miyagawa S, Takeo T, Nakagata N, Yamada G. Bmp4 is an essential growth factor for the initiation of genital tubercle (GT) outgrowth. Congenit Anom (Kyoto) 2020; 60:15-21. [PMID: 30714224 DOI: 10.1111/cga.12326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/31/2022]
Abstract
The external genitalia are appendage organs outgrowing from the posterior body trunk. Murine genital tubercle (GT), anlage of external genitalia, initiates its outgrowth from embryonic day (E) 10.5 as a bud structure. Several growth factors such as fibroblast growth factor (FGF), Wnt and Sonic hedgehog (Shh) are essential for the GT outgrowth. However, the mechanisms of initiation of GT outgrowth are poorly understood. We previously identified bone morphogenetic protein (Bmp) signaling as a negative regulator for GT outgrowth. We show here novel aspects of Bmp4 functions for GT outgrowth. We identified the Bmp4 was already expressed in cloaca region at E9.5, before GT outgrowth. To analyze the function of Bmp4 at early stage for the initiation of GT outgrowth, we utilized the Hoxa3-Cre driver and Bmp4 flox/flox mouse lines. Hoxa3 Cre/+ ; Bmp4 flox/flox mutant mice showed the hypoplasia of GT with reduced expression of outgrowth promoting genes such as Wnt5a, Hoxd13 and p63, whereas Shh expression was not affected. Formation of distal urethral epithelium (DUE) marked by the Fgf8 expression is essential for controlling mesenchymal genes expression in GT and subsequent its outgrowth. Furthermore, Fgf8 expression was dramatically reduced in such mutant mice indicating the defective DUE formation. Hence, current results indicate that Bmp4 is an essential growth factor for the initiation of GT outgrowth independent of Shh signaling. Thus, Bmp4 positively regulates for the formation of DUE. The current study provides new insights into the function of Bmp signaling at early stage for the initiation of GT outgrowth.
Collapse
Affiliation(s)
- Daiki Kajioka
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shoko Nakada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shoko Matsushita
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Miyagawa
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
17
|
Chen Y, Yu H, Pask AJ, Fujiyama A, Suzuki Y, Sugano S, Shaw G, Renfree MB. Hormone-responsive genes in the SHH and WNT/β-catenin signaling pathways influence urethral closure and phallus growth. Biol Reprod 2019; 99:806-816. [PMID: 29767687 DOI: 10.1093/biolre/ioy117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 05/13/2018] [Indexed: 11/14/2022] Open
Abstract
Environmental endocrine disruptors (EEDs) that affect androgen or estrogen activity may disrupt gene regulation during phallus development to cause hypospadias or a masculinized clitoris. We treated developing male tammar wallabies with estrogen and females with androgen from day 20-40 postpartum (pp) during the androgen imprinting window of sensitivity. Estrogen inhibited phallus elongation but had no effect on urethral closure and did not significantly depress testicular androgen synthesis. Androgen treatment in females did not promote phallus elongation but initiated urethral closure. Phalluses were collected for transcriptome sequencing at day 50 pp when they first become sexually dimorphic to examine changes in two signaling pathways, sonic hedgehog (SHH) and wingless-type MMTV integration site family (WNT)/β-catenin. SHH mRNA and β-catenin were predominantly expressed in the urethral epithelium in the tammar phallus, as in eutherian mammals. Estrogen treatment and castration of males induced an upregulation of SHH, while androgen treatment downregulated SHH. These effects appear to be direct since we detected putative estrogen receptor α (ERα) and androgen receptor (AR) binding sites near SHH. WNT5A, like SHH, was downregulated by androgen, while WNT4 was upregulated in female phalluses after androgen treatment. After estrogen treatment, WIF1 and WNT7A were both downregulated in male phalluses. After castration, WNT9A was upregulated. These results suggest that SHH and WNT pathways are regulated by both estrogen and androgen to direct the proliferation and elongation of the phallus during differentiation. Their response to exogenous hormones makes these genes potential targets of EEDs in the etiology of abnormal phallus development including hypospadias.
Collapse
Affiliation(s)
- Yu Chen
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Hongshi Yu
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Asao Fujiyama
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Sumio Sugano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Su T, Liu H, Zhang D, Xu G, Liu J, Evans SM, Pan J, Cui S. LIM homeodomain transcription factor Isl1 affects urethral epithelium differentiation and apoptosis via Shh. Cell Death Dis 2019; 10:713. [PMID: 31558700 PMCID: PMC6763423 DOI: 10.1038/s41419-019-1952-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/25/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Abstract
Urethral hypoplasia, including failure of urethral tube closure, is one of the common phenotypes observed in hereditary human disorders, the mechanism of which remains unclear. The present study was thus designed to study the expression, functions, and related mechanisms of the LIM homeobox transcription factor Isl1 throughout mouse urethral development. Results showed that Isl1 was highly expressed in urethral epithelial cells and mesenchymal cells of the genital tubercle (GT). Functional studies were carried out by utilizing the tamoxifen-inducible Isl1-knockout mouse model. Histological and morphological results indicated that Isl1 deletion caused urethral hypoplasia and inhibited maturation of the complex urethral epithelium. In addition, we show that Isl1-deleted mice failed to maintain the progenitor cell population required for renewal of urethral epithelium during tubular morphogenesis and exhibited significantly increased cell death within the urethra. Dual-Luciferase reporter assays and yeast one-hybrid assays showed that ISL1 was essential for normal urethral development by directly targeting the Shh gene. Collectively, results presented here demonstrated that Isl1 plays a crucial role in mouse urethral development, thus increasing our potential for understanding the mechanistic basis of hereditary urethral hypoplasia.
Collapse
Affiliation(s)
- Tiantian Su
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193, Beijing, People's Republic of China
| | - Hui Liu
- College of Veterinary Medicine, Yangzhou University, 225009, Yangzhou, Jiangsu, People's Republic of China
| | - Di Zhang
- College of Veterinary Medicine, Yangzhou University, 225009, Yangzhou, Jiangsu, People's Republic of China
| | - Guojin Xu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193, Beijing, People's Republic of China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193, Beijing, People's Republic of China
| | - Sylvia M Evans
- Skaggs School of Pharmacy, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jirong Pan
- Key Laboratory of Human Disease Comparative MedicineInstitute of Laboratory Animal Science, Chinese Academy of Medical Science and Comparative Medical Center, Peking Union Medical College, 100021, Beijing, People's Republic of China.
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, 100193, Beijing, People's Republic of China. .,College of Veterinary Medicine, Yangzhou University, 225009, Yangzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
19
|
Regulatory roles of epithelial-mesenchymal interaction (EMI) during early and androgen dependent external genitalia development. Differentiation 2019; 110:29-35. [PMID: 31590136 DOI: 10.1016/j.diff.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Development of external genitalia (ExG) has been a topic of long mystery in the field of organogenesis research. Early stage male and female of mouse embryos develop a common genital tubercle (GT) in the perineum whose outgrowth extends distally from the posterior cloacal regions. Concomitant with GT outgrowth, the cloaca is divided into urogenital sinus and anorectum by urorectal septum (URS) internally. The outgrowth of the GT is associated with the formation of endodermal epithelial urethral plate (UP) attached to the ventral epidermis of the GT. Such a common developmental phase is observed until around embryonic day 15.5 (E15.5) morphologically in mouse embryogenesis. Various growth factor genes, such as Fibroblast growth factor (Fgf) and Wnt genes are expressed and function during GT formation. Since the discovery of key growth factor signals and several regulatory molecules, elucidation of their functions has been achieved utilizing mouse developmental models, conditional gene knockout mouse and in vitro culture. Analyses on the phenotypes of such mouse models have revealed that several growth factor families play fundamental roles in ExG organogenesis based on the epithelial-mesenchymal interaction (EMI). More recently, EMI between developing urethral epithelia and its bilateral mesenchyme of later stages is also reported during subsequent stage of androgen-dependent male-type urethral formation in the mouse embryo. Mafb, belonging to AP-1 family and a key androgen-responsive mesenchymal gene, is identified and starts to be expressed around E14.5 when masculinization of the urethra is initiated. Mesenchymal cell condensation and migration, which are regulated by nonmuscle myosin, are shown to be essential process for masculinization. Hence, studies on EMI at various embryonic stages are important not only for early but also for subsequent masculinization of the urethra. In this review, a dynamic mode of EMI for both early and late phases of ExG development is discussed.
Collapse
|
20
|
Haller M, Ma L. Temporal, spatial, and genetic regulation of external genitalia development. Differentiation 2019; 110:1-7. [PMID: 31521888 DOI: 10.1016/j.diff.2019.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/26/2022]
Abstract
Fertilization requires the physical combination of gametes, and terrestrial mammals necessitated the evolution of genitalia capable of successfully completing the fertilization process in a non-aqueous environment. Thus, the male mammalian external genitalia evolved as an outgrowth from the body, an appendage sufficient to fertilize eggs housed deep inside the female. In this way, sexual dimorphism of mammalian genitalia became highly pronounced. This highly complex evolutionary divergence both from aqueous fertilization, as well as divergence between the sexes of terrestrial mammals, required exquisitely coordinated, novel patterns of gene expression to regulate the spatial and temporal events governing external genitalia development. Recent studies delineating the genetic regulation of external genitalia development, largely focusing on development of the murine genital tubercle, have vastly enlightened the field of reproductive developmental biology. Murine homologs of human genes have been selectively deleted in the mouse, either in the whole body or using tissue-specific and temporally-specific genetic drivers. The defects in outgrowth and urethral tubularization subsequent to the deletion of specific genes in the developing murine external genitalia delineates which genes are required in which compartments and at what times. This review details how these murine genetic models have created a somewhat modest but rapidly growing library of knowledge detailing the spatial-temporal genetic regulation of external genitalia development.
Collapse
Affiliation(s)
- Meade Haller
- Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA
| | - Liang Ma
- Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO, 63110, USA.
| |
Collapse
|
21
|
Sonic Hedgehog Signaling Is Required for Cyp26 Expression during Embryonic Development. Int J Mol Sci 2019; 20:ijms20092275. [PMID: 31072004 PMCID: PMC6540044 DOI: 10.3390/ijms20092275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023] Open
Abstract
Deciphering how signaling pathways interact during development is necessary for understanding the etiopathogenesis of congenital malformations and disease. In several embryonic structures, components of the Hedgehog and retinoic acid pathways, two potent players in development and disease are expressed and operate in the same or adjacent tissues and cells. Yet whether and, if so, how these pathways interact during organogenesis is, to a large extent, unclear. Using genetic and experimental approaches in the mouse, we show that during development of ontogenetically different organs, including the tail, genital tubercle, and secondary palate, Sonic hedgehog (SHH) loss-of-function causes anomalies phenocopying those induced by enhanced retinoic acid signaling and that SHH is required to prevent supraphysiological activation of retinoic signaling through maintenance and reinforcement of expression of the Cyp26 genes. Furthermore, in other tissues and organs, disruptions of the Hedgehog or the retinoic acid pathways during development generate similar phenotypes. These findings reveal that rigidly calibrated Hedgehog and retinoic acid activities are required for normal organogenesis and tissue patterning.
Collapse
|
22
|
Hashimoto D, Hyuga T, Acebedo AR, Alcantara MC, Suzuki K, Yamada G. Developmental mutant mouse models for external genitalia formation. Congenit Anom (Kyoto) 2019; 59:74-80. [PMID: 30554442 DOI: 10.1111/cga.12319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
Abstract
Development of external genitalia and perineum is the subject of developmental biology as well as toxicology and teratology researches. Cloaca forms in the lower (caudal) end of endoderm. Such endodermal epithelia and surrounding mesenchyme interact with various signals to form the external genitalia. External genitalia (the anlage termed as genital tubercle: GT) formation shows prominent sexually dimorphic morphogenesis in late embryonic stages, which is an unexplored developmental research field because of many reasons. External genitalia develop adjacent to the cloaca which develops urethra and corporal bodies. Developmental regulators including growth factor signals are necessary for epithelia-mesenchyme interaction (EMI) in posterior embryos including the cloaca and urethra in the genitalia. In the case of male type urethra, formation of tubular urethra proceeds from the lower (ventral) side of external genitalia as a masculinization process in contrast to the case of female urethra. Mechanisms for its development are not elucidated yet due to the lack of suitable mutant mouse models. Because of the recent progresses of Cre (recombinase)-mediated conditional target gene modification analyses, many developmental regulatory genes become increasingly analyzed. Conditional gene knockout mouse approaches and tissue lineage approaches are expected to offer vital information for such sexually dimorphic developmental processes. This review aims to offer recent updates on the progresses of these emerging developmental processes for the research field of congenital anomalies.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - Taiju Hyuga
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - Alvin R Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - Mellissa C Alcantara
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| |
Collapse
|
23
|
Ching ST, Infante CR, Du W, Sharir A, Park S, Menke DB, Klein OD. Isl1 mediates mesenchymal expansion in the developing external genitalia via regulation of Bmp4, Fgf10 and Wnt5a. Hum Mol Genet 2019; 27:107-119. [PMID: 29126155 DOI: 10.1093/hmg/ddx388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 10/25/2017] [Indexed: 12/20/2022] Open
Abstract
Genital malformations are among the most common human birth defects, and both genetic and environmental factors can contribute to these malformations. Development of the external genitalia in mammals relies on complex signaling networks, and disruption of these signaling pathways can lead to genital defects. Islet-1 (ISL1), a member of the LIM/Homeobox family of transcription factors, has been identified as a major susceptibility gene for classic bladder exstrophy in humans, a common form of the bladder exstrophy-epispadias complex (BEEC), and is implicated in a role in urinary tract development. We report that deletion of Isl1 from the genital mesenchyme in mice led to hypoplasia of the genital tubercle and prepuce, with an ectopic urethral opening and epispadias-like phenotype. These mice also developed hydroureter and hydronephrosis. Identification of ISL1 transcriptional targets via ChIP-Seq and expression analyses revealed that Isl1 regulates several important signaling pathways during embryonic genital development, including the BMP, WNT, and FGF cascades. An essential function of Isl1 during development of the external genitalia is to induce Bmp4-mediated apoptosis in the genital mesenchyme. Together, these studies demonstrate that Isl1 plays a critical role during development of the external genitalia and forms the basis for a greater understanding of the molecular mechanisms underlying the pathogenesis of BEEC and urinary tract defects in humans.
Collapse
Affiliation(s)
- Saunders T Ching
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA
| | - Carlos R Infante
- Department of Genetics, University of Georgia, GA 30602, USA.,Department of Molecular and Cellular Biology, University of Arizona, AZ 85721, USA
| | - Wen Du
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA.,State Key Laboratory of Oral Diseases, Department of Prosthetics, West China College of Stomatology, Sichuan University, Sichuan Sheng 610041, China
| | - Amnon Sharir
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA
| | - Sungdae Park
- Department of Genetics, University of Georgia, GA 30602, USA
| | - Douglas B Menke
- Department of Genetics, University of Georgia, GA 30602, USA
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143, USA.,Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Chen Y, Kuroki Y, Shaw G, Pask AJ, Yu H, Toyoda A, Fujiyama A, Renfree MB. Androgen and Oestrogen Affect the Expression of Long Non-Coding RNAs During Phallus Development in a Marsupial. Noncoding RNA 2018; 5:E3. [PMID: 30598023 PMCID: PMC6468475 DOI: 10.3390/ncrna5010003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 12/24/2022] Open
Abstract
There is increasing evidence that long non-coding RNAs (lncRNAs) are important for normal reproductive development, yet very few lncRNAs have been identified in phalluses so far. Unlike eutherians, phallus development in the marsupial tammar wallaby occurs post-natally, enabling manipulation not possible in eutherians in which differentiation occurs in utero. We treated with sex steroids to determine the effects of androgen and oestrogen on lncRNA expression during phallus development. Hormonal manipulations altered the coding and non-coding gene expression profile of phalluses. We identified several predicted co-regulatory lncRNAs that appear to be co-expressed with the hormone-responsive candidate genes regulating urethral closure and phallus growth, namely IGF1, AR and ESR1. Interestingly, more than 50% of AR-associated coding genes and lncRNAs were also associated with ESR1. In addition, we identified and validated three novel co-regulatory and hormone-responsive lncRNAs: lnc-BMP5, lnc-ZBTB16 and lncRSPO4. Lnc-BMP5 was detected in the urethral epithelium of male phalluses and was downregulated by oestrogen in males. Lnc-ZBTB16 was downregulated by oestrogen treatment in male phalluses at day 50 post-partum (pp). LncRSPO4 was downregulated by adiol treatment in female phalluses but increased in male phalluses after castration. Thus, the expression pattern and hormone responsiveness of these lncRNAs suggests a physiological role in the development of the phallus.
Collapse
Affiliation(s)
- Yu Chen
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Yoko Kuroki
- RIKEN, Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan.
| | - Geoff Shaw
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Hongshi Yu
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.
| | - Asao Fujiyama
- Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan.
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne 3010, VIC, Australia.
| |
Collapse
|
25
|
Ogino Y, Tohyama S, Kohno S, Toyota K, Yamada G, Yatsu R, Kobayashi T, Tatarazako N, Sato T, Matsubara H, Lange A, Tyler CR, Katsu Y, Iguchi T, Miyagawa S. Functional distinctions associated with the diversity of sex steroid hormone receptors ESR and AR. J Steroid Biochem Mol Biol 2018; 184:38-46. [PMID: 29885351 DOI: 10.1016/j.jsbmb.2018.06.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/26/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022]
Abstract
Sex steroid hormones including estrogens and androgens play fundamental roles in regulating reproductive activities and they act through estrogen and androgen receptors (ESR and AR). These steroid receptors have evolved from a common ancestor in association with several gene duplications. In most vertebrates, this has resulted in two ESR subtypes (ESR1 and ESR2) and one AR, whereas in teleost fish there are at least three ESRs (ESR1, ESR2a and ESR2b) and two ARs (ARα and ARβ) due to a lineage-specific whole genome duplication. Functional distinctions have been suggested among these receptors, but to date their roles have only been characterized in a limited number of species. Sexual differentiation and the development of reproductive organs are indispensable for all animal species and in vertebrates these events depend on the action of sex steroid hormones. Here we review the recent progress in understanding of the functions of the ESRs and ARs in the development and expression of sexually dimorphic characteristics associated with steroid hormone signaling in vertebrates, with representative fish, amphibians, reptiles, birds and mammals.
Collapse
Affiliation(s)
- Yukiko Ogino
- Attached Promotive Centre for International Education and Research of Agriculture, Faculty of Agriculture, Kyushu University, Fukuoka, Fukuoka 812-8581, Japan
| | - Saki Tohyama
- Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Shizuoka 422-8526, Japan
| | - Satomi Kohno
- Department of Biology, St. Cloud State University, St. Cloud, MN 56301, USA
| | - Kenji Toyota
- Department of Biological Sciences, Kanagawa University, Hiratsuka, Kanagawa 259-1293, Japan; Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Gen Yamada
- Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan
| | - Ryohei Yatsu
- Department of Integrative Biology, University of Texas at Austin, Austin, Texas 78712, USA
| | - Tohru Kobayashi
- Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Shizuoka 422-8526, Japan
| | | | - Tomomi Sato
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan
| | - Hajime Matsubara
- Department of Aquatic Biology, Faculty of Bioindustry, Tokyo University of Agriculture, Abashiri, Hokkaido 099-2493, Japan
| | - Anke Lange
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Charles R Tyler
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Yoshinao Katsu
- Graduate School of Life Science, Hokkaido University, Sapporo 060-0809, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan.
| | - Shinichi Miyagawa
- Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan; Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.
| |
Collapse
|
26
|
Regulation of masculinization: androgen signalling for external genitalia development. Nat Rev Urol 2018; 15:358-368. [DOI: 10.1038/s41585-018-0008-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Miyado M, Miyado K, Nakamura A, Fukami M, Yamada G, Oda SI. Expression patterns of Fgf8 and Shh in the developing external genitalia of Suncus murinus. Reproduction 2017; 153:187-195. [DOI: 10.1530/rep-16-0231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/23/2023]
Abstract
Reciprocal epithelial–mesenchymal interactions and several signalling pathways regulate the development of the genital tubercle (GT), an embryonic primordium of external genitalia. The morphology of the adult male external genitalia of the Asian house musk shrew Suncus murinus (hereafter, laboratory name: suncus) belonging to the order Eulipotyphla (the former order Insectivora or Soricomorpha) differs from those of mice and humans. However, the developmental process of the suncus GT and its regulatory genes are unknown. In the present study, we explored the morphological changes and gene expression patterns during the development of the suncus GT. Morphological observations suggested the presence of common (during the initial outgrowth) and species-specific (during the sexual differentiation of GT) developmental processes of the suncus GT. In gene expression analysis, fibroblast growth factor 8 (Fgf8) and sonic hedgehog (Shh), an indicator and regulator of GT development in mice respectively, were found to be expressed in the cloacal epithelium and the developing urethral epithelium of the suncus GT. This pattern of expression specifically in GT epithelium is similar to that observed in the developing mouse GT. Our results indicate that the mechanism of GT formation regulated by the FGF and SHH signalling pathways is widely conserved in mammals.
Collapse
|
28
|
Suzuki K, Matsumaru D, Matsushita S, Murashima A, Ludwig M, Reutter H, Yamada G. Epispadias and the associated embryopathies: genetic and developmental basis. Clin Genet 2016; 91:247-253. [PMID: 27649475 DOI: 10.1111/cge.12871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/25/2022]
Abstract
The abnormalities in the urogenital organs are frequently observed as human developmental diseases. Among such diseases, the defects in the upper part of external genitalia are rather rare named epispadias. The cleft in the dorsal part of external genitalia often reaches to the urethra. In general, the urogenital abnormalities accompany defects in the adjacent tissues and organs. The ventral body wall and bladder can also be affected in the patients with dorsal defects of the external genitalia. Therefore, such multiple malformations are often classified as bladder exstrophy and epispadias complex (BEEC). Because of the lower frequency of such birth defects and their early embryonic development, animal models are required to analyze the pathogenic mechanisms and the functions of responsible genes. Mutant mouse analyses on various signal cascades for external genitalia and body wall development are increasingly performed. The genetic interactions between growth factors such as bone morphogenetic proteins (Bmp) and transcription factors such as Msx1/2 and Isl1 have been suggested to play roles for such organogenesis. The significance of epithelial-mesenchymal interaction (EMI) is suggested during development. In this review, we describe on such local interactions and developmental regulators. We also introduce some mutant mouse models displaying external genitalia-body wall abnormalities.
Collapse
Affiliation(s)
- K Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - D Matsumaru
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - S Matsushita
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| | - A Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan.,Division of Human Embryology, Department of Anatomy, Iwate Medical University, Yahaba, Japan
| | - M Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University Hospital of Bonn, Bonn, Germany
| | - H Reutter
- Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany.,Department of Neonatology and Pediatric Intensive Care, University Hospital of Bonn, Bonn, Germany
| | - G Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University (WMU), Wakayama, Japan
| |
Collapse
|
29
|
Requirement for basement membrane laminin α5 during urethral and external genital development. Mech Dev 2016; 141:62-69. [PMID: 27208857 DOI: 10.1016/j.mod.2016.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 05/11/2016] [Accepted: 05/16/2016] [Indexed: 12/31/2022]
Abstract
Hypospadias, a congenital malformation of the penis characteristic of an abnormal urethral orifice, affects 1 in every 125 boys, and its incidence is rising. Herein we test the hypothesis that the basement membrane protein laminin α5 (LAMA5) plays a key role in the development of the mouse genital tubercle, the embryonic anlage of the external genitalia. Using standard histological analyses and electron microscopy, we characterized the morphology of the external genitalia in Lama5 knockout (LAMA5-KO) mouse embryos during both androgen-independent genital tubercle development and androgen-mediated sexual differentiation. We compared regulatory gene expression between control and LAMA5-KO by in situ hybridization. We also examined the epithelial structure of the mutant genital tubercle using immunofluorescence staining and histological analyses of semi-thin sections. We found that Lama5 was expressed in both ectodermal and endodermal epithelia of the cloaca. The LAMA5-KO displayed a profound external genital malformation in which the genital tubercle was underdeveloped with a large ectopic orifice at the proximal end. In older embryos, the urethra failed to form a tubular structure and was left completely exposed. These defects were not associated with a significant alteration in regulatory gene expression, but rather with a defective ectodermal epithelium and an abnormal disintegration of the cloacal membrane. We conclude that LAMA5 is required in the basement membrane to maintain normal architecture of the ventral ectoderm during genital tubercle development, which is essential for the formation of a tubular urethra. Perturbation of LAMA5, and possibly other basement membrane components, may cause hypospadias in humans.
Collapse
|
30
|
Armfield BA, Seifert AW, Zheng Z, Merton EM, Rock JR, Lopez MC, Baker HV, Cohn MJ. Molecular Characterization of the Genital Organizer: Gene Expression Profile of the Mouse Urethral Plate Epithelium. J Urol 2016; 196:1295-302. [PMID: 27173853 DOI: 10.1016/j.juro.2016.04.091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2016] [Indexed: 01/09/2023]
Abstract
PURPOSE Lower urinary tract malformations are among the most common congenital anomalies in humans. Molecular genetic studies of mouse external genital development have begun to identify mechanisms that pattern the genital tubercle and orchestrate urethral tubulogenesis. The urethral plate epithelium is an endodermal signaling region that has an essential role in external genital development. However, little is known about the molecular identity of this cell population or the genes that regulate its activity. MATERIALS AND METHODS We used microarray analysis to characterize differences in gene expression between urethral plate epithelium and surrounding tissue in mouse genital tubercles. In situ hybridizations were performed to map gene expression patterns and ToppCluster (https://toppcluster.cchmc.org/) was used to analyze gene associations. RESULTS A total of 84 genes were enriched at least 20-fold in urethral plate epithelium relative to surrounding tissue. The majority of these genes were expressed throughout the urethral plate in males and females at embryonic day 12.5 when the urethral plate is known to signal. Functional analysis using ToppCluster revealed genetic pathways with known functions in other organ systems but unknown roles in external genital development. Additionally, a 3-dimensional molecular atlas of genes enriched in urethral plate epithelium was generated and deposited at the GUDMAP (GenitoUrinary Development Molecular Anatomy Project) website (http://gudmap.org/). CONCLUSIONS We identified dozens of genes previously unknown to be expressed in urethral plate epithelium at a crucial developmental period. It provides a novel panel of genes for analysis in animal models and in humans with external genital anomalies.
Collapse
Affiliation(s)
- Brooke A Armfield
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Ashley W Seifert
- Department of Biology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Zhengui Zheng
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Emily M Merton
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Jason R Rock
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Henry V Baker
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida; Department of Biology, University of Florida Genetics Institute, University of Florida, Gainesville, Florida; Howard Hughes Medical Institute, University of Florida Genetics Institute, University of Florida, Gainesville, Florida.
| |
Collapse
|
31
|
Schultz NG, Ingels J, Hillhouse A, Wardwell K, Chang PL, Cheverud JM, Lutz C, Lu L, Williams RW, Dean MD. The Genetic Basis of Baculum Size and Shape Variation in Mice. G3 (BETHESDA, MD.) 2016; 6:1141-51. [PMID: 26935419 PMCID: PMC4856068 DOI: 10.1534/g3.116.027888] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/05/2016] [Indexed: 01/01/2023]
Abstract
The rapid divergence of male genitalia is a preeminent evolutionary pattern. This rapid divergence is especially striking in the baculum, a bone that occurs in the penis of many mammalian species. Closely related species often display diverse baculum morphology where no other morphological differences can be discerned. While this fundamental pattern of evolution has been appreciated at the level of gross morphology, nearly nothing is known about the genetic basis of size and shape divergence. Quantifying the genetic basis of baculum size and shape variation has been difficult because these structures generally lack obvious landmarks, so comparing them in three dimensions is not straightforward. Here, we develop a novel morphometric approach to quantify size and shape variation from three-dimensional micro-CT scans taken from 369 bacula, representing 75 distinct strains of the BXD family of mice. We identify two quantitative trait loci (QTL) that explain ∼50% of the variance in baculum size, and a third QTL that explains more than 20% of the variance in shape. Together, our study demonstrates that baculum morphology may diverge relatively easily, with mutations at a few loci of large effect that independently modulate size and shape. Based on a combination of bioinformatic investigations and new data on RNA expression, we prioritized these QTL to 16 candidate genes, which have hypothesized roles in bone morphogenesis and may enable future genetic manipulation of baculum morphology.
Collapse
Affiliation(s)
- Nicholas G Schultz
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - Jesse Ingels
- University of Tennessee, Health Science Center, Memphis, Tennessee 38163
| | - Andrew Hillhouse
- Texas A & M, Veterinary Medicine and Biomedical Sciences, College Station, Texas 77845
| | | | - Peter L Chang
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| | - James M Cheverud
- Loyola University, Department of Biology, Chicago, Illinois 60626
| | | | - Lu Lu
- University of Tennessee, Health Science Center, Memphis, Tennessee 38163
| | - Robert W Williams
- University of Tennessee, Health Science Center, Memphis, Tennessee 38163
| | - Matthew D Dean
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
32
|
Investigation of sexual dimorphisms through mouse models and hormone/hormone-disruptor treatments. Differentiation 2016; 91:78-89. [DOI: 10.1016/j.diff.2015.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 11/11/2015] [Indexed: 01/23/2023]
|
33
|
Reutter H, Keppler-Noreuil K, E Keegan C, Thiele H, Yamada G, Ludwig M. Genetics of Bladder-Exstrophy-Epispadias Complex (BEEC): Systematic Elucidation of Mendelian and Multifactorial Phenotypes. Curr Genomics 2016; 17:4-13. [PMID: 27013921 PMCID: PMC4780475 DOI: 10.2174/1389202916666151014221806] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
The Bladder-Exstrophy-Epispadias Complex (BEEC) represents the severe end of the uro-rectal malformation spectrum, and has a profound impact on continence, and on sexual and renal function. While previous reports of familial occurrence, in-creased recurrence among first-degree relatives, high concordance rates among monozygotic twins, and chromosomal aberra-tions were suggestive of causative genetic factors, the recent identification of copy number variations (CNVs), susceptibility regions and genes through the systematic application of array based analysis, candidate gene and genome-wide association studies (GWAS) provide strong evidence. These findings in human BEEC cohorts are underscored by the recent description of BEEC(-like) murine knock-out models. Here, we discuss the current knowledge of the potential molecular mechanisms, mediating abnormal uro-rectal development leading to the BEEC, demonstrating the importance of ISL1-pathway in human and mouse and propose SLC20A1 and CELSR3 as the first BEEC candidate genes, identified through systematic whole-exome sequencing (WES) in BEEC patients.
Collapse
Affiliation(s)
- Heiko Reutter
- Department of Neonatology and Pediatric Intensive Care; Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Kim Keppler-Noreuil
- Human Development Section, National Human Genome Research Institute, Bethesda, MD, USA
| | - Catherine E Keegan
- Department of Pediatric Genetics, University of Michigan Medical Center, Michigan, USA
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Gen Yamada
- Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Japan
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| |
Collapse
|
34
|
Anderson MJ, Southon E, Tessarollo L, Lewandoski M. Fgf3-Fgf4-cis: A new mouse line for studying Fgf functions during mouse development. Genesis 2016; 54:91-8. [PMID: 26666435 DOI: 10.1002/dvg.22913] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 01/15/2023]
Abstract
The fibroblast growth factor (FGF) family consists of 22 ligands in mice and humans. FGF signaling is vital for embryogenesis and, when dysregulated, can cause disease. Loss-of-function genetic analysis in the mouse has been crucial for understanding FGF function. Such analysis has revealed that multiple Fgfs sometimes function redundantly. Exploring such redundancy between Fgf3 and Fgf4 is currently impossible because both genes are located on chromosome 7, about 18.5 kb apart, making the frequency of interallelic cross-over between existing mutant alleles too infrequent to be practicable. Therefore, we retargeted Fgf3 and Fgf4 in cis, generating an Fgf3 null allele and a conditional Fgf4 allele, subject to Cre inactivation. To increase the frequency of cis targeting, we used an F1 embryonic stem cell line that contained 129/SvJae (129) and C57BL/6J (B6) chromosomes and targeting constructs isogenic to the 129 chromosome. We confirmed cis targeting by assaying for B6/129 allele-specific single-nucleotide polymorphisms. We demonstrated the utility of the Fgf3(Δ)-Fgf4(flox)-cis mouse line by showing that the caudal axis extension defects found in the Fgf3 mutants worsen when Fgf4 is also inactivated. This Fgf3(Δ)-Fgf4(flox)-cis line will be useful to study redundancy of these genes in a variety of tissues and stages in development.
Collapse
Affiliation(s)
- Matthew J Anderson
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, 21702, Maryland
| | - Eileen Southon
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, 21702, Maryland
| | - Lino Tessarollo
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, 21702, Maryland
| | - Mark Lewandoski
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, 21702, Maryland
| |
Collapse
|
35
|
Timing of androgen receptor disruption and estrogen exposure underlies a spectrum of congenital penile anomalies. Proc Natl Acad Sci U S A 2015; 112:E7194-203. [PMID: 26598695 DOI: 10.1073/pnas.1515981112] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Congenital penile anomalies (CPAs) are among the most common human birth defects. Reports of CPAs, which include hypospadias, chordee, micropenis, and ambiguous genitalia, have risen sharply in recent decades, but the causes of these malformations are rarely identified. Both genetic anomalies and environmental factors, such as antiandrogenic and estrogenic endocrine disrupting chemicals (EDCs), are suspected to cause CPAs; however, little is known about the temporal window(s) of sensitivity to EDCs, or the tissue-specific roles and downstream targets of the androgen receptor (AR) in external genitalia. Here, we show that the full spectrum of CPAs can be produced by disrupting AR at different developmental stages and in specific cell types in the mouse genital tubercle. Inactivation of AR during a narrow window of prenatal development results in hypospadias and chordee, whereas earlier disruptions cause ambiguous genitalia and later disruptions cause micropenis. The neonatal phase of penile development is controlled by the balance of AR to estrogen receptor α (ERα) activity; either inhibition of androgen or augmentation of estrogen signaling can induce micropenis. AR and ERα have opposite effects on cell division, apoptosis, and regulation of Hedgehog, fibroblast growth factor, bone morphogenetic protein, and Wnt signaling in the genital tubercle. We identify Indian hedgehog (Ihh) as a novel downstream target of AR in external genitalia and show that conditional deletion of Ihh inhibits penile masculinization. These studies reveal previously unidentified cellular and molecular mechanisms by which antiandrogenic and estrogenic signals induce penile malformations and demonstrate that the timing of endocrine disruption can determine the type of CPA.
Collapse
|
36
|
Clarification of mammalian cloacal morphogenesis using high-resolution episcopic microscopy. Dev Biol 2015; 409:106-113. [PMID: 26485363 DOI: 10.1016/j.ydbio.2015.10.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 10/07/2015] [Accepted: 10/15/2015] [Indexed: 01/03/2023]
Abstract
The developmental process through which the cloaca transforms from one hollow structure to two separated urinary and digestive outlets remains controversial and speculative. Here, we use high-resolution episcopic microscopy to examine a comprehensive series of normal and mutant mouse cloaca in which the detailed 3-dimensional (3-D) morphological features are illuminated throughout the development. We provide evidence that the dorsal peri-cloacal mesenchyme (dPCM) remains stationary while other surrounding tissues grow towards it. This causes dramatic changes of spatial relationship among caudal structures and morphological transformation of the cloaca. The 3-D characterizations of Dkk1 mutants reveal a hyperplastic defect of dPCM, which leads to a significant anterior shift of the caudal boundary of the cloaca, premature occlusion of the cloaca and, imperforate anus phenotype. Conversely, Shh knockout causes a severe hypoplastic defect of cloaca mesenchyme including dPCM and persistent cloaca. Collectively, these findings suggest that formation of the dPCM is critical for cloacal morphogenesis and furthermore, growth and movement of the mesenchymal tissues towards the dPCM lead to the cloaca occlusion and separation of the urinary and digestive outlets.
Collapse
|
37
|
Infante CR, Mihala AG, Park S, Wang JS, Johnson KK, Lauderdale JD, Menke DB. Shared Enhancer Activity in the Limbs and Phallus and Functional Divergence of a Limb-Genital cis-Regulatory Element in Snakes. Dev Cell 2015; 35:107-19. [PMID: 26439399 DOI: 10.1016/j.devcel.2015.09.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 07/24/2015] [Accepted: 09/09/2015] [Indexed: 11/18/2022]
Abstract
The amniote phallus and limbs differ dramatically in their morphologies but share patterns of signaling and gene expression in early development. Thus far, the extent to which genital and limb transcriptional networks also share cis-regulatory elements has remained unexplored. We show that many limb enhancers are retained in snake genomes, suggesting that these elements may function in non-limb tissues. Consistent with this, our analysis of cis-regulatory activity in mice and Anolis lizards reveals that patterns of enhancer activity in embryonic limbs and genitalia overlap heavily. In mice, deletion of HLEB, an enhancer of Tbx4, produces defects in hindlimbs and genitalia, establishing the importance of this limb-genital enhancer for development of these different appendages. Further analyses demonstrate that the HLEB of snakes has lost hindlimb enhancer function while retaining genital activity. Our findings identify roles for Tbx4 in genital development and highlight deep similarities in cis-regulatory activity between limbs and genitalia.
Collapse
Affiliation(s)
- Carlos R Infante
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | | | - Sungdae Park
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Jialiang S Wang
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Kenji K Johnson
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - James D Lauderdale
- Department of Genetics, University of Georgia, Athens, GA 30602, USA; Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
38
|
Systematic stereoscopic analyses for cloacal development: The origin of anorectal malformations. Sci Rep 2015; 5:13943. [PMID: 26354024 PMCID: PMC4564729 DOI: 10.1038/srep13943] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/05/2015] [Indexed: 12/28/2022] Open
Abstract
The division of the embryonic cloaca is the most essential event for the formation of digestive and urinary tracts. The defective development of the cloaca results in anorectal malformations (ARMs; 2–5 per 10,000 live births). However, the developmental and pathogenic mechanisms of ARMs are unclear. In the current study, we visualized the epithelia in the developing cloaca and nephric ducts (NDs). Systemic stereoscopic analyses revealed that the ND-cloaca connection sites shifted from the lateral-middle to dorsal-anterior part of the cloaca during cloacal division from E10.5 to E11.5 in mouse embryos. Genetic cell labeling analyses revealed that the cells in the ventral cloacal epithelium in the early stages rarely contributed to the dorsal part. Moreover, we revealed the possible morphogenetic movement of endodermal cells within the anterior part of the urogenital sinus and hindgut. These results provide the basis for understanding both cloacal development and the ARM pathogenesis.
Collapse
|
39
|
Abbo O, Ferdynus C, Kalfa N, Huiart L, Sauvat F, Harper LH. Male infants with hypospadias and/or cryptorchidism show a lower 2D/4D digit ratio than normal boys. Arch Dis Child 2015; 100:643-7. [PMID: 25688099 DOI: 10.1136/archdischild-2014-306454] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 01/26/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND In humans the ratio of the index finger to the ring finger is sexually dimorphic, with the mean ratio being larger in women than in men. It has been suggested that this difference is related to prenatal androgen exposure. This has been further demonstrated in children with congenital adrenal hyperplasia. Normal development of the male external genitalia is linked to androgen-mediated events during gestation. We therefore wanted to determine if the 2D:4D digit ratio was normal in boys with cryptorchidism or hypospadias. METHODS We prospectively enrolled all prepubertal patients seen in the outpatient clinic for cryptorchidism or hypospadias between September and December 2012. We then compared their 2D:4D digit ratio with two control groups made up of normal boys and normal girls. Interobserver and intraobserver variability was evaluated. RESULTS We included 57 boys with hypospadias and/or cryptorchidism, 79 boys without genital abnormalities and 25 girls without genital abnormalities. The mean 2D:4D ratio for both hands was significantly different between the three groups, with the digit ratio for boys with genital anomalies being lower than for normal boys and normal girls (p<0.0001). CONCLUSIONS It appears that boys with genital abnormalities (cryptorchidism and/or hypospadias) have a lower 2D:4D digit ratio than boys without genital anomalies.
Collapse
Affiliation(s)
- O Abbo
- Department of Pediatric Surgery, CHU F Guyon, Bellepierre, Saint-Denis de La Réunion, Reunion Island, France
| | - C Ferdynus
- Unité de Soutien Méthodologique, CHU F Guyon, Bellepierre, Saint-Denis de La Réunion, Reunion Island, France
| | - N Kalfa
- Department of Pediatric Surgery, Hôpital Lapeyronie, CHU Montpellier, Montpellier, France
| | - L Huiart
- Unité de Soutien Méthodologique, CHU F Guyon, Bellepierre, Saint-Denis de La Réunion, Reunion Island, France
| | - F Sauvat
- Department of Pediatric Surgery, CHU F Guyon, Bellepierre, Saint-Denis de La Réunion, Reunion Island, France
| | - L H Harper
- Department of Pediatric Surgery, CHU F Guyon, Bellepierre, Saint-Denis de La Réunion, Reunion Island, France
| |
Collapse
|
40
|
Harada M, Omori A, Nakahara C, Nakagata N, Akita K, Yamada G. Tissue-specific roles of FGF signaling in external genitalia development. Dev Dyn 2015; 244:759-73. [DOI: 10.1002/dvdy.24277] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/22/2015] [Accepted: 03/22/2015] [Indexed: 11/11/2022] Open
Affiliation(s)
- Masayo Harada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Akiko Omori
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| | - Chiaki Nakahara
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering; Center for Animal Resources and Development, Kumamoto University; Kumamoto Japan
| | - Keiichi Akita
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Gen Yamada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| |
Collapse
|
41
|
Sequencing of the DKK1 gene in patients with anorectal malformations and hypospadias. Eur J Pediatr 2015; 174:583-7. [PMID: 25319845 DOI: 10.1007/s00431-014-2436-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 01/08/2023]
Abstract
UNLABELLED Anorectal malformations (ARM) are rare congenital malformations of the gastrointestinal tract. Approximately 60% of the patients have additional congenital malformations, such as hypospadias. A recently published article showed that deletion of one single gene, dickkopf WNT signaling pathway inhibitor-1 (Dkk1), resulted in an imperforate anus with rectourinary fistula and preputial hypospadias in mice. To determine whether DKK1 also plays a role in the etiology of ARM and hypospadias in humans, we sequenced the four exons of the DKK1 gene in 17 patients affected with both ARM and hypospadias. No new potential disease-causing variant was identified. However, we detected a known non-synonymous variant in one patient, which was predicted in silico to be damaging, and the corresponding unaffected amino acid is highly conserved. CONCLUSION In this human study, a potential interesting non-synonymous variant was found in the DKK1 gene. Whether this variant plays a contributory role in the genesis of ARM or hypospadias would require a much larger study.
Collapse
|
42
|
Tschopp P, Sherratt E, Sanger TJ, Groner AC, Aspiras AC, Hu JK, Pourquié O, Gros J, Tabin CJ. A relative shift in cloacal location repositions external genitalia in amniote evolution. Nature 2014; 516:391-4. [PMID: 25383527 PMCID: PMC4294627 DOI: 10.1038/nature13819] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 08/20/2014] [Indexed: 12/23/2022]
Abstract
The move of vertebrates to a terrestrial lifestyle required major adaptations in their locomotory apparatus and reproductive organs. While the fin-to-limb transition has received considerable attention, little is known about the developmental and evolutionary origins of external genitalia. Similarities in gene expression have been interpreted as a potential evolutionary link between the limb and genitals; however, no underlying developmental mechanism has been identified. We re-examined this question using micro-computed tomography, lineage tracing in three amniote clades, and RNA-sequencing-based transcriptional profiling. Here we show that the developmental origin of external genitalia has shifted through evolution, and in some taxa limbs and genitals share a common primordium. In squamates, the genitalia develop directly from the budding hindlimbs, or the remnants thereof, whereas in mice the genital tubercle originates from the ventral and tail bud mesenchyme. The recruitment of different cell populations for genital outgrowth follows a change in the relative position of the cloaca, the genitalia organizing centre. Ectopic grafting of the cloaca demonstrates the conserved ability of different mesenchymal cells to respond to these genitalia-inducing signals. Our results support a limb-like developmental origin of external genitalia as the ancestral condition. Moreover, they suggest that a change in the relative position of the cloacal signalling centre during evolution has led to an altered developmental route for external genitalia in mammals, while preserving parts of the ancestral limb molecular circuitry owing to a common evolutionary origin.
Collapse
Affiliation(s)
- Patrick Tschopp
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Emma Sherratt
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138
| | - Thomas J. Sanger
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138
| | - Anna C. Groner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115
| | - Ariel C. Aspiras
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Jimmy K. Hu
- Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
| | - Jérôme Gros
- Developmental and Stem Cell Biology Department, Institut Pasteur, 75724 Paris Cedex 15, France
| | | |
Collapse
|
43
|
Lonfat N, Montavon T, Darbellay F, Gitto S, Duboule D. Convergent evolution of complex regulatory landscapes and pleiotropy at Hox loci. Science 2014; 346:1004-6. [DOI: 10.1126/science.1257493] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
44
|
Sexually dimorphic expression of Mafb regulates masculinization of the embryonic urethral formation. Proc Natl Acad Sci U S A 2014; 111:16407-12. [PMID: 25362053 DOI: 10.1073/pnas.1413273111] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Masculinization of external genitalia is an essential process in the formation of the male reproductive system. Prominent characteristics of this masculinization are the organ size and the sexual differentiation of the urethra. Although androgen is a pivotal inducer of the masculinization, the regulatory mechanism under the control of androgen is still unknown. Here, we address this longstanding question about how androgen induces masculinization of the embryonic external genitalia through the identification of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb) gene. Mafb is expressed prominently in the mesenchyme of male genital tubercle (GT), the anlage of external genitalia. MAFB expression is rarely detected in the mesenchyme of female GTs. However, exposure to exogenous androgen induces its mesenchymal expression in female GTs. Furthermore, MAFB expression is prominently down-regulated in male GTs of androgen receptor (Ar) KO mice, indicating that AR signaling is necessary for its expression. It is revealed that Mafb KO male GTs exhibit defective embryonic urethral formation, giving insight into the common human congenital anomaly hypospadias. However, the size of Mafb KO male GTs is similar with that of wild-type males. Moreover, androgen treatment fails to induce urethral masculinization of the GTs in Mafb KO mice. The current results provide evidence that Mafb is an androgen-inducible, sexually dimorphic regulator of embryonic urethral masculinization.
Collapse
|
45
|
Zeidler C, Woelfle J, Draaken M, Mughal SS, Große G, Hilger AC, Dworschak GC, Boemers TM, Jenetzky E, Zwink N, Lacher M, Schmidt D, Schmiedeke E, Grasshoff-Derr S, Märzheuser S, Holland-Cunz S, Schäfer M, Bartels E, Keppler K, Palta M, Leonhardt J, Kujath C, Rißmann A, Nöthen MM, Reutter H, Ludwig M. Heterozygous FGF8 mutations in patients presenting cryptorchidism and multiple VATER/VACTERL features without limb anomalies. ACTA ACUST UNITED AC 2014; 100:750-9. [PMID: 25131394 DOI: 10.1002/bdra.23278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/31/2014] [Accepted: 06/03/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND The acronym VATER/VACTERL association describes the combination of at least three of the following cardinal features: vertebral defects, anorectal malformations, cardiac defects, tracheoesophageal fistula with or without esophageal atresia, renal malformations, and limb defects. Although fibroblast growth factor-8 (FGF8) mutations have mainly found in patients with Kallmann syndrome, mice with a hypomorphic Fgf8 allele or complete gene invalidation display, aside from gonadotropin-releasing hormone deficiency, parts or even the entire spectrum of human VATER/VACTERL association. METHODS We performed FGF8 gene analysis in 49 patients with VATER/VACTERL association and 27 patients presenting with a VATER/VACTERL-like phenotype (two cardinal features). RESULTS We identified two heterozygous FGF8 mutations in patients displaying either VATER/VACTERL association (p.Gly29_Arg34dup) or a VATER/VACTERL-like phenotype (p.Pro26Leu) without limb anomalies. Whereas the duplication mutation has not been reported before, p.Pro26Leu was once observed in a Kallmann syndrome patient. Both our patients had additional bilateral cryptorchidism, a key phenotypic feature in males with FGF8 associated Kallmann syndrome. Each mutation was paternally inherited. Besides delayed puberty in both and additional unilateral cryptorchidism in one of the fathers, they were otherwise healthy. Serum hormone levels downstream the gonadotropin-releasing hormone in both patients and their fathers were within normal range. CONCLUSION Our results suggest FGF8 mutations to contribute to the formation of the VATER/VACTERL association. Further studies are needed to support this observation.
Collapse
Affiliation(s)
- Claudia Zeidler
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gredler ML, Seifert AW, Cohn MJ. Morphogenesis and Patterning of the Phallus and Cloaca in the American Alligator, Alligator mississippiensis. Sex Dev 2014; 9:53-67. [DOI: 10.1159/000364817] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
47
|
Leal F, Cohn MJ. Development of Hemipenes in the Ball Python Snake Python regius. Sex Dev 2014; 9:6-20. [DOI: 10.1159/000363758] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
48
|
Gredler ML, Sanger TJ, Cohn MJ. Development of the Cloaca, Hemipenes, and Hemiclitores in the Green Anole, Anolis carolinensis. Sex Dev 2014; 9:21-33. [DOI: 10.1159/000363757] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
49
|
Murashima A, Kishigami S, Thomson A, Yamada G. Androgens and mammalian male reproductive tract development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:163-70. [PMID: 24875095 DOI: 10.1016/j.bbagrm.2014.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/28/2014] [Accepted: 05/19/2014] [Indexed: 12/31/2022]
Abstract
One of the main functions of androgen is in the sexually dimorphic development of the male reproductive tissues. During embryogenesis, androgen determines the morphogenesis of male specific organs, such as the epididymis, seminal vesicle, prostate and penis. Despite the critical function of androgens in masculinization, the downstream molecular mechanisms of androgen signaling are poorly understood. Tissue recombination experiments and tissue specific androgen receptor (AR) knockout mouse studies have revealed epithelial or mesenchymal specific androgen-AR signaling functions. These findings also indicate that epithelial-mesenchymal interactions are a key feature of AR specific activity, and paracrine growth factor action may mediate some of the effects of androgens. This review focuses on mouse models showing the interactions of androgen and growth factor pathways that promote the sexual differentiation of reproductive organs. Recent studies investigating context dependent AR target genes are also discussed. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Aki Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan
| | - Satoshi Kishigami
- Faculty of Biology-Oriented Science and Technology, Kinki University, Kinokawa 649-6493, Wakayama, Japan
| | - Axel Thomson
- Department of Urology, McGill University Health Centre, 1650 Cedar Av, Montreal, Québec, H3A 1A4, Canada
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan.
| |
Collapse
|
50
|
Ng RCL, Matsumaru D, Ho ASH, Garcia-Barceló MM, Yuan ZW, Smith D, Kodjabachian L, Tam PKH, Yamada G, Lui VCH. Dysregulation of Wnt inhibitory factor 1 (Wif1) expression resulted in aberrant Wnt-β-catenin signaling and cell death of the cloaca endoderm, and anorectal malformations. Cell Death Differ 2014; 21:978-89. [PMID: 24632949 PMCID: PMC4013516 DOI: 10.1038/cdd.2014.20] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/29/2013] [Accepted: 11/13/2013] [Indexed: 02/08/2023] Open
Abstract
In mammalian urorectal development, the urorectal septum (urs) descends from the ventral body wall to the cloaca membrane (cm) to partition the cloaca into urogenital sinus and rectum. Defective urs growth results in human congenital anorectal malformations (ARMs), and their pathogenic mechanisms are unclear. Recent studies only focused on the importance of urs mesenchyme proliferation, which is induced by endoderm-derived Sonic Hedgehog (Shh). Here, we showed that the programmed cell death of the apical urs and proximal cm endoderm is particularly crucial for the growth of urs during septation. The apoptotic endoderm was closely associated with the tempo-spatial expression of Wnt inhibitory factor 1 (Wif1), which is an inhibitor of Wnt-β-catenin signaling. In Wif1lacZ/lacZ mutant mice and cultured urorectum with exogenous Wif1, cloaca septation was defective with undescended urs and hypospadias-like phenotypes, and such septation defects were also observed in Shh−/− mutants and in endodermal β-catenin gain-of-function (GOF) mutants. In addition, Wif1 and Shh were expressed in a complementary manner in the cloaca endoderm, and Wif1 was ectopically expressed in the urs and cm associated with excessive endodermal apoptosis and septation defects in Shh−/− mutants. Furthermore, apoptotic cells were markedly reduced in the endodermal β-catenin GOF mutant embryos, which counteracted the inhibitory effects of Wif1. Taken altogether, these data suggest that regulated expression of Wif1 is critical for the growth of the urs during cloaca septation. Hence, Wif1 governs cell apoptosis of urs endoderm by repressing β-catenin signal, which may facilitate the protrusion of the underlying proliferating mesenchymal cells towards the cm for cloaca septation. Dysregulation of this endodermal Shh-Wif1-β-catenin signaling axis contributes to ARM pathogenesis.
Collapse
Affiliation(s)
- R C-L Ng
- 1] Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China [2] Centre of Reproduction, Development and Growth, Hong Kong SAR, China
| | - D Matsumaru
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - A S-H Ho
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - M-M Garcia-Barceló
- 1] Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China [2] Centre of Reproduction, Development and Growth, Hong Kong SAR, China
| | - Z-W Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shengyang, China
| | - D Smith
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - L Kodjabachian
- Aix-Marseille Université CNRS UMR 7288, Institut de Biologie du Dévelopment de Marseille, Marseille, France
| | - P K-H Tam
- 1] Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China [2] Centre of Reproduction, Development and Growth, Hong Kong SAR, China
| | - G Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - V C-H Lui
- 1] Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China [2] Centre of Reproduction, Development and Growth, Hong Kong SAR, China
| |
Collapse
|