1
|
Yoshida S, Yoshida K. Regulatory mechanisms governing GLI proteins in hedgehog signaling. Anat Sci Int 2025; 100:143-154. [PMID: 39576500 DOI: 10.1007/s12565-024-00814-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/14/2024] [Indexed: 02/16/2025]
Abstract
The Hedgehog (Hh) signaling pathway is critical for regulating cell growth, survival, fate determination, and the overall patterning of both vertebrate and invertebrate body plans. Aberrations in Hh signaling are associated with congenital abnormalities and tumorigenesis. In vertebrates, Hh signaling depends uniquely on primary cilia, microtubule-based organelles that extend from the cell surface. Over the last 2 decades, studies have demonstrated that key molecules regulating Hh signaling dynamically accumulate in primary cilia via intraflagellar transport systems. Moreover, through the primary cilia, extracellular signals are converted to stabilize GLI2 and GLI3 that are transcription factors that play a central role in regulating Hh signaling at the post-translational modification level. Recent in vivo and anatomical studies have uncovered crucial molecules that facilitate the conversion of extracellular signals into the intracellular stabilization of GLI2/GLI3 via primary cilia, emphasizing their essential roles in tissue development and tumorigenesis. This review explores the regulatory mechanisms of GLI2/GLI3 with a focus on mammalian tissue development.
Collapse
Affiliation(s)
- Saishu Yoshida
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, 274-8510, Japan.
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| |
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 2025; 254:102-141. [PMID: 39319771 PMCID: PMC11809137 DOI: 10.1002/dvdy.735] [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: 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 SciencesUniversity of HertfordshireHatfieldUK
| | - Bashar Aldeiri
- Department of Paediatric SurgeryChelsea and Westminster HospitalLondonUK
| | - Mark Davenport
- Department of Paediatric SurgeryKing's College HospitalLondonUK
| | | |
Collapse
|
3
|
van Bever Y, Boers RG, Brüggenwirth HT, van IJcken WF, Magielsen FJ, de Klein A, Boers JB, Looijenga LH, Brosens E, Gribnau J, Hannema SE. Genome-wide methylation analysis in patients with proximal hypospadias - a pilot study and review of the literature. Epigenetics 2024; 19:2392048. [PMID: 39151125 PMCID: PMC11373573 DOI: 10.1080/15592294.2024.2392048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/03/2024] [Accepted: 08/06/2024] [Indexed: 08/18/2024] Open
Abstract
In patients with proximal hypospadias, often no genetic cause is identified despite extensive genetic testing. Many genes involved in sex development encode transcription factors with strict timing and dosing of the gene products. We hypothesised that there might be recurrent differences in DNA methylation in boys with hypospadias and that these might differ between patients born small versus appropriate for gestational age. Genome-wide Methylated DNA sequencing (MeD-seq) was performed on 32bp LpnPI restriction enzyme fragments after RE-digestion in leucocytes from 16 XY boys with unexplained proximal hypospadias, one with an unexplained XX testicular disorder/difference of sex development (DSD) and twelve, healthy, sex- and age-matched controls. Five of seven differentially methylated regions (DMRs) between patients and XY controls were in the Long Intergenic Non-Protein Coding RNA 665 (LINC00665; CpG24525). Three patients showed hypermethylation of MAP3K1. Finally, no DMRs in XX testicular DSD associated genes were identified in the XX boy versus XX controls. In conclusion, we observed no recognizable epigenetic signature in 16 boys with XY proximal hypospadias and no difference between children born small versus appropriate for gestational age. Comparison to previous methylation studies in individuals with hypospadias did not show consistent findings, possibly due to the use of different inclusion criteria, tissues and methods.
Collapse
Affiliation(s)
- Yolande van Bever
- Department of Clinical Genetics, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
- Erasmus MC Center of Expertise for DSD, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Ruben G Boers
- Department of Developmental Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Hennie T Brüggenwirth
- Department of Clinical Genetics, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
- Erasmus MC Center of Expertise for DSD, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Wilfred Fj van IJcken
- Center for Biomics, Erasmus MC, Rotterdam, The Netherlands
- Department of Cell biology, Erasmus MC, Rotterdam, Netherlands
| | - Frank J Magielsen
- Department of Clinical Genetics, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Joachim B Boers
- Department of Developmental Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Leendert Hj Looijenga
- Princess Máxima Center for Pediatric Oncology, Utrecht, and Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Erwin Brosens
- Department of Clinical Genetics, Erasmus MC, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Sabine E Hannema
- Erasmus MC Center of Expertise for DSD, Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Paediatric Endocrinology, Amsterdam University Medical Centers, Location Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Gavazzi LM, Nair M, Suydam R, Usip S, Thewissen JGM, Cooper LN. Protein signaling and morphological development of the tail fluke in the embryonic beluga whale (Delphinapterus leucas). Dev Dyn 2024; 253:859-874. [PMID: 38494595 PMCID: PMC11656686 DOI: 10.1002/dvdy.704] [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: 04/21/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND During the land-to-sea transition of cetaceans (whales, dolphins, and porpoises), the hindlimbs were lost and replaced by an elaborate tail fluke that evolved 32 Ma. All modern cetaceans utilize flukes for lift-based propulsion, and nothing is known of this organ's molecular origins during embryonic development. This study utilizes immunohistochemistry to identify the spatiotemporal location of protein signals known to drive appendage outgrowth in other vertebrates (e.g., Sonic Hedgehog [SHH], GREMLIN [GREM], wingless-type family member 7a [WNT], and fibroblast growth factors [FGFs]) and to test the hypothesis that signals associated with outgrowth and patterning of the tail fluke are similar to a tetrapod limb. Specifically, this study utilizes an embryo of a beluga whale (Delphinapterus leucas) as a case study. RESULTS Results showed epidermal signals of WNT and FGFs, and mesenchymal/epidermal signals of SHH and GREM. These patterns are most consistent with vertebrate limb development. Overall, these data are most consistent with the hypothesis that outgrowth of tail flukes in cetaceans employs a signaling pattern that suggests genes essential for limb outgrowth and patterning shape this evolutionarily novel appendage. CONCLUSIONS While these data add insights into the molecular signals potentially driving the evolution and development of tail flukes in cetaceans, further exploration of the molecular drivers of fluke development is required.
Collapse
Affiliation(s)
- L. M. Gavazzi
- School of Biomedical SciencesKent State UniversityKentOhioUSA
- Musculoskeletal Research Focus Area, Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - M. Nair
- Wright State UniversityDaytonOhioUSA
| | - R. Suydam
- Department of Wildlife ManagementNorth Slope BoroughUtqiaġvikAlaskaUSA
| | - S. Usip
- Musculoskeletal Research Focus Area, Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - J. G. M. Thewissen
- Musculoskeletal Research Focus Area, Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| | - L. N. Cooper
- Musculoskeletal Research Focus Area, Department of Anatomy and NeurobiologyNortheast Ohio Medical UniversityRootstownOhioUSA
| |
Collapse
|
5
|
Fendereski K, Schaeffer AJ. Bladder exstrophy-epispadias-cloacal exstrophy complex: characteristics, aetiologies, and epidemiologic findings. AFRICAN UROLOGY 2024; 4:S20-S25. [PMID: 39687282 PMCID: PMC11649331 DOI: 10.36303/auj.0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Bladder exstrophy-epispadias-cloacal exstrophy complex (BEEC) is a spectrum of congenital urologic anomalies that involve the bladder, urethra, genitalia, and pelvic musculoskeletal system, and can affect urinary continence, sexual health, and fertility. BEEC includes a wide spectrum of anatomical abnormalities with different levels of severity: epispadias represents the mildest phenotype, classic bladder exstrophy (CBE) is the most common defect, and cloacal exstrophy (CE) - often referred to as omphalocele, exstrophy, imperforate anus, and spinal defects (OEIS) complex - is the most severe form. BEEC disorders cause significant health problems and affect the health-related quality of life (QoL) of affected individuals. There have been significant insights into the aetiology of BEEC in the last decade. Specifically, recent genetic studies have suggested that downstream regulator(s) of p63, Isl1, and other genes may play a role in the failure of the lower urinary tract to close. This narrative review outlines the unique anatomy of bladder exstrophy (BE) and epispadias, with a brief mention of the anatomy found in CE. A literature review using PubMed and Google Scholar databases was used to identify relevant articles on the outlined topics without placing any limitations on publication years or study designs. We included full-text English articles published in peer-reviewed journals related to the terms: "exstrophy" & "epispadias" AND "aetiology", "embryology" and "incidence". We summarise the epidemiology of this rare complex - including what is known about its incidence in Africa - before presenting recent advances in comparative genetics from mouse models and human studies that provide insights into BEEC pathogenesis.
Collapse
Affiliation(s)
- K Fendereski
- Division of Urology, Department of Surgery, University of Utah School of Medicine, United States of America
| | - A J Schaeffer
- Division of Urology, Department of Surgery, University of Utah School of Medicine, United States of America
- Intermountain Primary Children's Hospital, United States of America
| |
Collapse
|
6
|
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: 3] [Impact Index Per Article: 3.0] [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
|
7
|
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
|
8
|
Olson CM, Frolov A, Tan Y, Martin JR, Campbell M. A Rare Case of Penoscrotal Webbing and Extensive Hernias: An Anatomical Report With Genetic Insights. Cureus 2023; 15:e47375. [PMID: 38021525 PMCID: PMC10657503 DOI: 10.7759/cureus.47375] [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] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
During a routine anatomical dissection of an 81-year-old male cadaver received through the Gift Body Program of Saint Louis University School of Medicine (SLU SOM), a massive bulging in the abdominal area was observed that was consistent with numerous hernia repairs noted in the donor's self-reported medical history. Gross anatomical dissection of the cadaveric body revealed extensive herniation of portions of the small intestine and peritoneal sac along the costal margin and extending to the left aspect of the abdomen. Additionally, an uncircumcised phallus was buried within the suprapubic fat pad and demonstrated simple, grade III penoscrotal webbing (PSW), creating an impression of micropenis presence. To gain additional insights into the current case, analysis of the coding regions (exomes) of DNA procured from the body for putative genetic variants was performed using next-generation sequencing (NGS) technology. This analysis revealed 110 rare (minor allele frequency (MAF) ≤ 0.01), pathologic/deleterious genetic mutations. The most relevant variants to this case were the ones associated with male sexual development, BMP1 and BMP4; connective tissue development, COL3A1 and COL5A3; cilia morphogenesis and function, DNAH5 and MAPK15; as well as hormonal homeostasis, ESR1. Direct involvement of BMP1 both in male sexual development and hernia genesis makes it a strong candidate for linking the two pathologies, PSW and multiple hernias, observed in the present case. Yet the presence of a group of mutated genes linked to myopathies (ITGA7, NRAP, POLM, SCN5A, XIRP2) and muscular dystrophy (ITGA7) raises a question about the involvement of these muscular pathologies in hernia genesis and unsuccessful hernia repairs associated with the current case.
Collapse
Affiliation(s)
- Carley M Olson
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, Saint Louis, USA
| | - Andrey Frolov
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, Saint Louis, USA
| | - Yun Tan
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, Saint Louis, USA
| | - John R Martin
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, Saint Louis, USA
| | - Meadow Campbell
- Department of Surgery - Center for Anatomical Science and Education, Saint Louis University School of Medicine, Saint Louis, USA
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
Fujimoto K, Hashimoto D, Kashimada K, Kumegawa S, Ueda Y, Hyuga T, Hirashima T, Inoue N, Suzuki K, Hara I, Asamura S, Yamada G. A visualization system for erectile vascular dynamics. Front Cell Dev Biol 2022; 10:1000342. [PMID: 36313553 PMCID: PMC9615422 DOI: 10.3389/fcell.2022.1000342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Erection is an essential process which requires the male penis for copulation. This copulatory process depends on the vascular dynamic regulation of the penis. The corpus cavernosum (CC) in the upper (dorsal) part of the penis plays a major role in regulating blood flow inside the penis. When the CC is filled with blood, the sinusoids, including micro-vessels, dilate during erection. The CC is an androgen-dependent organ, and various genital abnormalities including erectile dysfunction (ED) are widely known. Previous studies have shown that androgen deprivation by castration results in significantly decreased smooth muscles of the CC. Experimental works in erectile biology have previously measured intracavernosal penile pressure and mechanical tension. Such reports analyze limited features without assessing the dynamic aspects of the erectile process. In the current study, we established a novel explant system enabling direct visual imaging of the sinusoidal lumen to evaluate the dynamic movement of the cavernous space. To analyze the alternation of sinusoidal spaces, micro-dissected CC explants by patent blue dye injection were incubated and examined for their structural alternations during relaxation/contraction. The dynamic process of relaxation/contraction was analyzed with various external factors administered to the CC. The system enabled the imaging of relaxation/contraction of the lumens of the sinusoids and the collagen-containing tissues. Histological analysis on the explant system also showed the relaxation/contraction. Thus, the system mimics the regulatory process of dynamic relaxation/contraction in the erectile response. The current system also enabled evaluating the erectile pathophysiology. In the current study, the lumen of sinusoids relaxed/contracted in castrated mice similarly with normal mice. These results suggested that the dynamic erectile relaxation/contraction process was similarly retained in castrated mice. However, the system also revealed decreased duration time of erection in castrated mice. The current study is expected to promote further understanding of the pathophysiology of ED, which will be useful for new treatments in the future. Hence, the current system provides unique information to investigate the novel regulations of erectile function, which can provide tools for analyzing the pathology of ED.
Collapse
Affiliation(s)
- Kota Fujimoto
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
- Department of Plastic and Reconstructive Surgery, Wakayama Medical University, Wakayama, Japan
| | - Daiki Hashimoto
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Kumegawa
- Department of Plastic and Reconstructive Surgery, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ueda
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Taiju Hyuga
- Department of Pediatric Urology, Children’s Medical Center Tochigi, Jichi Medical University, Tochigi, Japan
| | - Tsuyoshi Hirashima
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Kentaro Suzuki
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi, Japan
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive Surgery, Wakayama Medical University, Wakayama, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
- Department of Plastic and Reconstructive Surgery, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Gen Yamada,
| |
Collapse
|
13
|
McClelland K, Li W, Rosenblum ND. Pallister-Hall syndrome, GLI3, and kidney malformation. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:264-278. [PMID: 36165461 DOI: 10.1002/ajmg.c.31999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/06/2022] [Accepted: 08/27/2022] [Indexed: 01/29/2023]
Abstract
Pallister-Hall syndrome (PHS) is a rare autosomal dominant disease diagnosed by the presence of hypothalamic hamartoma, mesoaxial polydactyly and a truncating variant in the middle third of the GLI-Kruppel family member 3 (GLI3) gene. PHS may also include a wide range of clinical phenotypes affecting multiple organ systems including congenital anomalies of the kidney and urinary tract (CAKUT). The observed clinical phenotypes are consistent with the essential role of GLI3, a transcriptional effector in the hedgehog (Hh) signaling pathway, in organogenesis. However, the mechanisms by which truncation of GLI3 in PHS results in such a variety of clinical phenotypes with variable severity, even within the same organ, remain unclear. In this study we focus on presentation of CAKUT in PHS. A systematic analysis of reported PHS patients (n = 78) revealed a prevalence of 26.9% (21/78) of CAKUT. Hypoplasia (± dysplasia) and agenesis were the two main types of CAKUT; bilateral and unilateral CAKUT were reported with equal frequency. Examination of clinical phenotypes with CAKUT revealed a significant association between CAKUT and craniofacial defects, bifid epiglottis and a Disorder of Sex Development, specifically affecting external genitalia. Lastly, we determined that PHS patients with CAKUT predominately had substitution type variants (as opposed to deletion type variants in non-CAKUT PHS patients) in the middle third of the GLI3 gene. These results provide a foundation for future work aimed at uncovering the molecular mechanisms by which variant GLI3 result in the wide range and severity of clinical features observed in PHS.
Collapse
Affiliation(s)
- Kathryn McClelland
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Weili Li
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Norman D Rosenblum
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Nephrology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Hashimoto D, Fujimoto K, Morioka S, Ayabe S, Kataoka T, Fukumura R, Ueda Y, Kajimoto M, Hyuga T, Suzuki K, Hara I, Asamura S, Wakana S, Yoshiki A, Gondo Y, Tamura M, Sasaki T, Yamada G. Establishment of mouse line showing inducible priapism-like phenotypes. Reprod Med Biol 2022; 21:e12472. [PMID: 35765371 PMCID: PMC9207557 DOI: 10.1002/rmb2.12472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/11/2022] Open
Abstract
Purpose Penile research is expected to reveal new targets for treatment and prevention of the complex mechanisms of its disorder including erectile dysfunction (ED). Thus, analyses of the molecular processes of penile ED and continuous erection as priapism are essential issues of reproductive medicine. Methods By performing mouse N-ethyl-N-nitrosourea mutagenesis and exome sequencing, we established a novel mouse line displaying protruded genitalia phenotype (PGP; priapism-like phenotype) and identified a novel Pitpna gene mutation for PGP. Extensive histological analyses on the Pitpna mutant and intracavernous pressure measurement (ICP) and liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI/MS)/MS analyses were performed. Results We evaluated the role of phospholipids during erection for the first time and showed the mutants of inducible phenotypes of priapism. Moreover, quantitative analysis using LC-ESI/MS/MS revealed that the level of phosphatidylinositol (PI) was significantly lower in the mutant penile samples. These results imply that PI may contribute to penile erection by PITPα. Conclusions Our findings suggest that the current mutant is a mouse model for priapism and abnormalities in PI signaling pathways through PITPα may lead to priapism providing an attractive novel therapeutic target in its treatment.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shin Morioka
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Shinya Ayabe
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Kataoka
- Department of Clinical PharmaceuticsGraduate School of Medical SciencesNagoya City UniversityNagoyaJapan
| | - Ryutaro Fukumura
- Clinical Laboratories Department sSRL & Shizuoka Cancer Center Collaborative Laboratories, IncShizuoka PrefJapan
| | - Yuko Ueda
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Mizuki Kajimoto
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Taiju Hyuga
- Department of Pediatric UrologyChildren's Medical Center TochigiJichi Medical UniversityTochigiJapan
| | - Kentaro Suzuki
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Isao Hara
- Department of UrologyWakayama Medical UniversityWakayamaJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shigeharu Wakana
- Department of Animal ExperimentationFoundation for Biomedical Research and Innovation at KobeCreative Lab for Innovation in Kobe 5F 6‐3‐7KobeHyogoJapan
| | - Atsushi Yoshiki
- Experimental Animal DivisionRIKEN BioResource Research CenterIbarakiJapan
| | - Yoichi Gondo
- Department of Molecular Life SciencesDivision of Basic Medical Science and Molecular MedicineTokai University School of MedicineIsehara‐shiKanagawaJapan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype AnalysisRIKEN BioResource Research CenterTsukubaIbarakiJapan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology/Lipid BiologyMedical Research InstituteTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Gen Yamada
- Department of Developmental GeneticsInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
15
|
Murugapoopathy V, Cammisotto PG, Mossa AH, Campeau L, Gupta IR. Osr1 Is Required for Mesenchymal Derivatives That Produce Collagen in the Bladder. Int J Mol Sci 2021; 22:ijms222212387. [PMID: 34830270 PMCID: PMC8619163 DOI: 10.3390/ijms222212387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/24/2022] Open
Abstract
The extracellular matrix of the bladder consists mostly of type I and III collagen, which are required during loading. During bladder injury, there is an accumulation of collagen that impairs bladder function. Little is known about the genes that regulate production of collagens in the bladder. We demonstrate that the transcription factor Odd-skipped related 1 (Osr1) is expressed in the bladder mesenchyme and epithelium at the onset of development. As development proceeds, Osr1 is mainly expressed in mesenchymal progenitors and their derivatives. We hypothesized that Osr1 regulates mesenchymal cell differentiation and production of collagens in the bladder. To test this hypothesis, we examined newborn and adult mice heterozygous for Osr1, Osr1+/−. The bladders of newborn Osr1+/− mice had a decrease in collagen I by western blot analysis and a global decrease in collagens using Sirius red staining. There was also a decrease in the cellularity of the lamina propria, where most collagen is synthesized. This was not due to decreased proliferation or increased apoptosis in this cell population. Surprisingly, the bladders of adult Osr1+/− mice had an increase in collagen that was associated with abnormal bladder function; they also had a decrease in bladder capacity and voided more frequently. The results suggest that Osr1 is important for the differentiation of mesenchymal cells that give rise to collagen-producing cells.
Collapse
Affiliation(s)
| | - Philippe G. Cammisotto
- Lady Davis Research Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (P.G.C.); (A.H.M.); (L.C.)
| | - Abubakr H. Mossa
- Lady Davis Research Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (P.G.C.); (A.H.M.); (L.C.)
| | - Lysanne Campeau
- Lady Davis Research Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (P.G.C.); (A.H.M.); (L.C.)
- Division of Urology, Department of Surgery, Jewish General Hospital, McGill University, Montreal, QCH3T 1E2, Canada
| | - Indra R. Gupta
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada;
- Research Institute of the McGill University Health Center, Montreal, QC H3H 2R9, Canada
- Department of Pediatrics, McGill University, Montreal, QC H4A 3J1, Canada
- Correspondence:
| |
Collapse
|
16
|
Alcantara MC, Suzuki K, Acebedo AR, Sakamoto Y, Nishita M, Minami Y, Kikuchi A, Yamada G. Stage-dependent function of Wnt5a during male external genitalia development. Congenit Anom (Kyoto) 2021; 61:212-219. [PMID: 34255394 DOI: 10.1111/cga.12438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
External genitalia development in mice involves multiple developmental processes under the regulation of various signaling pathways. Wnt5a, one of the major Wnt ligands, is a crucial developmental regulator of outgrowing organs such as the limb, the mandible, and the external genitalia. Defects in Wnt5a signaling have been linked to Robinow syndrome, a genetic disorder in which male patients manifest a micropenis and defective urethral tube formation. Whereas Wnt5a is required for cell proliferation during embryonic external genitalia outgrowth, its role for urethral tube formation has yet to be understood. Here, we show that Wnt5a contributes to urethral tube formation as well as external genitalia outgrowth. Wnt5a is expressed in the embryonic external genitalia mesenchyme, and mesenchymal-specific conditional Wnt5a knockout mice resulted in hypospadias-like urethral defects. Early deletion of Wnt5a at E10.5 showed severe defects in both external genitalia outgrowth and urethral tube formation, along with reduced cell proliferation. The severe urethral tube defect persisted during later timing deletion of Wnt5a (E13.5). Further analyses revealed that loss of Wnt5a disrupted cell polarity and led to a reduction of the phosphorylated myosin light chain and the focal adhesion protein, vinculin. Altogether, these results suggest that Wnt5a coordinates cell proliferation and directed cell migration in a stage-dependent manner during male external genitalia development. Furthermore, Wnt5a may regulate cell polarity, focal adhesion formation, and cell contractility, leading to directed cell migration during male-type urethral formation in a manner that has not been reported in other organ fusion events.
Collapse
Affiliation(s)
- Mellissa C Alcantara
- 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
| | - Alvin R Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuki Sakamoto
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Michiru Nishita
- Department of Biochemistry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuhiro Minami
- Faculty of Medical Sciences, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
17
|
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: 7] [Impact Index Per Article: 1.8] [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
|
18
|
Saraç M, Canpolat Ş, Önalan Etem E, Tektemur A, Tartar T, Bakal U, Kazez A. The role of sonic hedgehog homologue signal pathway in hypospadias aetiology. J Pediatr Urol 2021; 17:630.e1-630.e7. [PMID: 34275739 DOI: 10.1016/j.jpurol.2021.06.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Hypospadias is one of the most common congenital anomalies of the male genitalia. Sonic hedgehog homologue (SHH) signalling pathway is believed to be involved in the development of the male genital system. OBJECTIVE In this clinical prospective study, the role of the SHH pathway in hypospadias aetiology was investigated. STUDY DESIGN In this study, 200 healthy children (boys without hypospadias, control group), 118 patients (boys with distal hypospadias) and 82 patients (boys with proximal hypospadias) of age 0-16 years were included. The expression of the genes suppressor of fused protein (SUFU), SHH, protein patched homologue (PTCH; PTCH1 and PTCH2), glioma-associated oncogene homologue (GLI; GLI1, GLI2, GLI3 and GLI4), smoothened, frizzled-class receptor (SMO) and serine/threonine-protein kinase 36 (STK36) that are involved in SHH pathway were investigated. Furthermore, polymorphism analyses of GLI2, SHH and PTCH1 genes were performed. The history of hypospadias in the first and second-degree relatives of the patients in boys with distal hypospadias and boys with proximal hypospadias was inquired. RESULTS Ten patients in the boys with distal hypospadias and twenty patients in the boys with proximal hypospadias had a history of hypospadias in first or second-degree relatives (p < 0.05). There was a significant decrease in mRNA expressions of SHH and PTCH1 genes in boys with proximal hypospadias compared to boys without hypospadias (p < 0.05). Besides, a significant decrease in mRNA fold-change of GLI2 gene was detected in boys with both distal hypospadias and proximal hypospadias compared to boys without hypospadias (p < 0.05). In contrast, there was no significant difference in the mRNA fold-changes of PTCH2, SUFU, GLI1, GLI3, GLI4, SMO and STK36 genes among the groups. Moreover, there were no significant differences in the frequencies of variant genotypes and alleles rs735557, rs12711538 and rs4848632 (GLI2 gene), rs104894049 (SHH gene) and rs41313327 (PTCH1 gene) (p > 0.05). DISCUSSION SHH expression is required for the growth and differentiation of the genital bulge. Developmental defects in the external genital organs were demonstrated in mice with SHH deletion. It has been demonstrated that SHH mainly plays a role in the formation of sinusoid morphology of the penis. In the present study, although SHH and PTCH gene expressions were found to be decreased only in the penile tissues of proximal hypospadias, GLI2 gene expression was decreased in penile tissues of boys with both distal hypospadias and boys with proximal hypospadias. CONCLUSION Genes involved in the SHH pathway might play a role in the aetiology of hypospadias. Furthermore, there is a correlation between molecular defects in this pathway and severity of hypospadias.
Collapse
Affiliation(s)
- Mehmet Saraç
- Department of Pediatric Surgery, Firat University School of Medicine, Elazig, 23119, Turkey.
| | - Şenay Canpolat
- Ministry of Health University, Ankara Dr. Sami Ulus Obstetrics, Child Health and Diseases Training and Research Hospital, Elazig, 23119, Turkey.
| | - Ebru Önalan Etem
- Department of Medical Biology, Firat University School of Medicine, 23119, Elazig, Turkey.
| | - Ahmet Tektemur
- Department of Medical Biology, Firat University School of Medicine, 23119, Elazig, Turkey.
| | - Tugay Tartar
- Department of Pediatric Surgery, Firat University School of Medicine, Elazig, 23119, Turkey.
| | - Unal Bakal
- Department of Pediatric Surgery, Firat University School of Medicine, Elazig, 23119, Turkey.
| | - Ahmet Kazez
- Department of Pediatric Surgery, Firat University School of Medicine, Elazig, 23119, Turkey.
| |
Collapse
|
19
|
Female congenital aphallia: a unique case of congenital absence of the clitoris with an ectopic labium majorum. Clin Dysmorphol 2021; 30:17-21. [PMID: 33136658 DOI: 10.1097/mcd.0000000000000354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The spectrum of disorders of sexual development includes anatomical abnormalities of the external genitalia, the phenotypic variability of which and the underlying causes are numerous. However, female aphallia and ectopia of the labium majorum appear to be some of the rarest forms of external genitalia malformations. Aphallia is mostly described in males with a frequency of less than one per 40 000 male newborns. Although syndromic forms of aphallia in females have been reported, for example, in Robinow, CHARGE, and Prader-Willi syndrome, reports of isolated female aphallia are meager. Here, we describe the first case of isolated agenesis of the clitoris with an ectopic labium majorum and review the literature of this uncommon malformation and its potential dysmorphogenetic mechanism. We emphasize the need for a routine exhaustive physical examination to identify and characterize this unusual malformation correctly so that families can be appropriately counseled as to cause and potential complications.
Collapse
|
20
|
Kothandapani A, Jefcoate CR, Jorgensen JS. Cholesterol Contributes to Male Sex Differentiation Through Its Developmental Role in Androgen Synthesis and Hedgehog Signaling. Endocrinology 2021; 162:6204698. [PMID: 33784378 PMCID: PMC8168945 DOI: 10.1210/endocr/bqab066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 12/17/2022]
Abstract
Two specialized functions of cholesterol during fetal development include serving as a precursor to androgen synthesis and supporting hedgehog (HH) signaling activity. Androgens are produced by the testes to facilitate masculinization of the fetus. Recent evidence shows that intricate interactions between the HH and androgen signaling pathways are required for optimal male sex differentiation and defects of either can cause birth anomalies indicative of 46,XY male variations of sex development (VSD). Further, perturbations in cholesterol synthesis can cause developmental defects, including VSD, that phenocopy those caused by disrupted androgen or HH signaling, highlighting the functional role of cholesterol in promoting male sex differentiation. In this review, we focus on the role of cholesterol in systemic androgen and local HH signaling events during fetal masculinization and their collective contributions to pediatric VSD.
Collapse
Affiliation(s)
- Anbarasi Kothandapani
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Correspondence: Anbarasi Kothandapani, PhD, Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53705, USA. E-mail:
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Correspondence: Joan S. Jorgensen, DVM, PhD, Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53705, USA. E-mail:
| |
Collapse
|
21
|
Armfield BA, Cohn MJ. Single cell transcriptomic analysis of external genitalia reveals complex and sexually dimorphic cell populations in the early genital tubercle. Dev Biol 2021; 477:145-154. [PMID: 34033822 DOI: 10.1016/j.ydbio.2021.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 11/27/2022]
Abstract
External genital organs are among the most recognizable sexually dimorphic characters. The penis and clitoris develop from the embryonic genital tubercle, an outgrowth at the anterior margin of the cloaca that undergoes an extensive period of development in male and female embryos prior to the onset of sexual differentiation. In mice, differentiation into the penis and clitoris begins around embryonic day (E)15.5. Current knowledge of cell types that comprise the genital tubercle is limited to a few studies that have fate mapped derivatives of endoderm, mesoderm, and ectoderm. Here we use single cell transcriptomics to characterize the cell populations in the genital tubercles of male and female mouse embryos at E14.5, approximately 24 h before the onset of sexual differentiation, and we present the first comprehensive atlas of single-cell gene expression during external genital development. Clustering analyses and annotation using marker genes shows 19 distinct cell populations in E14.5 genital tubercles. Mapping of cell clusters to anatomical locations using in situ gene expression patterns revealed granularity of cellular specializations and positional identities. Although E14.5 precedes sexually dimorphic morphogenesis of the genital tubercle, comparative analysis of males and females identified sexual dimorphisms at the single cell level, including male-specific cell clusters with transcriptional signatures of smooth muscle and bone progenitors, both of which are known to be sexually dimorphic in adult genitalia, as well as immune cells. These results provide a new resource for classification of external genital cell types based on gene expression profiles and reveal sex-specific cellular specializations in the early genital tubercle.
Collapse
Affiliation(s)
- Brooke A Armfield
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA; Department of Biology, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
22
|
Pollini D, Loffredo R, Maniscalco F, Cardano M, Micaelli M, Bonomo I, Licata NV, Peroni D, Tomaszewska W, Rossi A, Crippa V, Dassi E, Viero G, Quattrone A, Poletti A, Conti L, Provenzani A. Multilayer and MATR3-dependent regulation of mRNAs maintains pluripotency in human induced pluripotent stem cells. iScience 2021; 24:102197. [PMID: 33733063 PMCID: PMC7940987 DOI: 10.1016/j.isci.2021.102197] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/22/2020] [Accepted: 02/11/2021] [Indexed: 10/28/2022] Open
Abstract
Matrin3 (MATR3) is a nuclear RNA/DNA-binding protein that plays pleiotropic roles in gene expression regulation by directly stabilizing target RNAs and supporting the activity of transcription factors by modulating chromatin architecture. MATR3 is involved in the differentiation of neural cells, and, here, we elucidate its critical functions in regulating pluripotent circuits in human induced pluripotent stem cells (hiPSCs). MATR3 downregulation affects hiPSCs' differentiation potential by altering key pluripotency regulators' expression levels, including OCT4, NANOG, and LIN28A by pleiotropic mechanisms. MATR3 binds to the OCT4 and YTHDF1 promoters favoring their expression. YTHDF1, in turn, binds the m6A-modified OCT4 mRNA. Furthermore, MATR3 is recruited on ribosomes and controls pluripotency regulating the translation of specific transcripts, including NANOG and LIN28A, by direct binding and favoring their stabilization. These results show that MATR3 orchestrates the pluripotency circuitry by regulating the transcription, translational efficiency, and epitranscriptome of specific transcripts.
Collapse
Affiliation(s)
- Daniele Pollini
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Rosa Loffredo
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Federica Maniscalco
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
- Institute of Biophysics, CNR, Trento, Italy
| | - Marina Cardano
- Cell Technology Core Facility, Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Mariachiara Micaelli
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Isabelle Bonomo
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Daniele Peroni
- Mass Spectrometry Core Facility, Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Weronika Tomaszewska
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Annalisa Rossi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Valeria Crippa
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Erik Dassi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Angelo Poletti
- Laboratorio di Biologia Applicata, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luciano Conti
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
23
|
Harsono M, Yanishevski D, Pourcyrous M. Congenital Perineal Groove Defect in Monozygotic Twin Infants: A Literature Review. AJP Rep 2021; 11:e54-e57. [PMID: 33815912 PMCID: PMC8012879 DOI: 10.1055/s-0041-1727145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 10/26/2022] Open
Abstract
Perineal groove is a rare benign congenital anomaly with lesion that resembles perforation of mid-perineum or perineal raphe area. Most reported cases of congenital perineal groove presented as an isolated defect in term or early-term singleton female infants. Thus far, there is no reported case of this anomaly in monozygotic twins. Embryo pathogenesis of this female predominance congenital defect remains controversial. Many clinicians are unfamiliar with this congenital anomaly. This congenital defect tends to get self-resolved at around 2 year of age. Nevertheless, the exposed nonepithelized mucous membrane can carry risk of local infection or irritation with the possibility of requiring early surgical correction. The defect can be infrequently associated with other ano-urogenital malformations that required immediate surgical intervention. Most isolated cases tend to be asymptomatic and self-healed with expectant management. Surgical correction may be considered if not healed after 2 years of age. Early diagnosis at birth is important to avoid misdiagnoses at later age for trauma, dermatitis, sexual abuse, and risk of unnecessary aggressive intervention. Early parental counseling for providing good hygiene and close follow-up is important to prevent infection or inflammation. Presentation of this anomaly in both monozygotic twins may support the hypothesis of potential disruption during embryo morphogenesis stages.
Collapse
Affiliation(s)
- Mimily Harsono
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - David Yanishevski
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Massroor Pourcyrous
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Obstetrics/Gynecology, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
24
|
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
|
25
|
Prenatal diagnosis and neonatal phenotype of a de novo microdeletion of 17p11.2p12 associated with Smith‒Magenis syndrome and external genital defects. J Genet 2020. [DOI: 10.1007/s12041-020-01213-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
26
|
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
|
27
|
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
|
28
|
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: 14] [Impact Index Per Article: 2.3] [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
|
29
|
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
|
30
|
Schilbach-Rott syndrome associated with 9q22.32q22.33 duplication, involving the PTCH1 gene. Eur J Hum Genet 2019; 27:1260-1266. [PMID: 30936464 DOI: 10.1038/s41431-019-0385-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 03/05/2019] [Accepted: 03/12/2019] [Indexed: 11/08/2022] Open
Abstract
Schilbach-Rott syndrome (SRS, OMIM%164220) is a disorder of unknown aetiology that is characterised by hypotelorism, epichantal folds, cleft palate, dysmorphic face, hypospadia in males and mild mental retardation in some patients. To date, 5 families and 17 patients have exhibited this phenotype, and recurrence in two of these families suggests an autosomal dominant inheritance. SRS overlaps with a mild form of holoprosencephaly (HPE), but array-CGH analysis and sequencing of some HPE-related genes (SEPT9, SHH and TWIST) did not reveal any variants in at least one family. Herein, we investigated by array-CGH analysis a 11-year-old female patient and her father, both exhibiting the typical SRS phenotype, disclosing in the daughter-father couple the same microduplication of chromosome 9q22.32q22.33 [arr[hg19]9q22.32(98,049,611_98,049,636)x3,9q22.33 (99,301,483_99,301,508)x3], involving eight genes, including PTCH1. The duplication segregated with the disease, since it was not found in the healthy paternal grandparents of the proband. The gain-of-function variants of the PTCH1 gene are responsible for a mild form of HPE. This is the first genetic variant found in SRS. This finding reinforces the hypothesis that SRS belongs to the HPE clinical spectrum and suggests to perform array-CGH in patients with SRS phenotype and, if negative, to consider a potential benefit from sequencing of HPE-related genes.
Collapse
|
31
|
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
|
32
|
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: 44] [Impact Index Per Article: 6.3] [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
|
33
|
A Rare Case of Human Diphallia Associated with Hypospadias. Case Rep Urol 2018; 2018:8293036. [PMID: 30009078 PMCID: PMC6020512 DOI: 10.1155/2018/8293036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/24/2018] [Indexed: 11/18/2022] Open
Abstract
Diphallia or penile duplication is a rare congenital variant with an estimated frequency of 1 per 5 to 6 million live births. The extent of duplication varies widely and typically occurs with other malformations including urogenital, gastrointestinal, and musculoskeletal anomalies. Here we present a case of human diphallia that was detected during routine dissection of an 84-year-old cadaver. Upon thorough examination, this case was characterized as a complete bifid penis which was accompanied by hypospadias with no other anatomical abnormalities detected. To gain insights into the etiology of this case, we analyzed DNA procured from the body for putative genetic variants using Next Generation Sequencing (NGS) technology. Our results support clinical observations consistent with human diphallia being a polygenic syndrome and identify new genetic variants that might underlie its etiology.
Collapse
|
34
|
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]
|
35
|
Zhou Y, Liu X, Huang F, Liu Y, Cao X, Shen L, Long C, He D, Lin T, Wei G. Epithelial-mesenchymal transformation and apoptosis in rat urethra development. Pediatr Res 2017; 82:1073-1079. [PMID: 28876330 DOI: 10.1038/pr.2017.185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/24/2017] [Indexed: 01/04/2023]
Abstract
BackgroundTo examine the mechanism of urethral seam formation during embryonal development of rat urethra.MethodsTime-mated Sprague-Dawley rats were killed and the genital tubercles of male pups harvested on embryonic day (ED) 15, 16, 18, and 19. External morphology was observed under scanning electron microscope. Serial transverse sections were prepared to examine dynamic changes in the urethral seam morphology with hematoxylin-eosin staining, immunohistochemistry, transmission electron microscopy, and double immunofluorescence.ResultsBilateral outgrowth of urethral swelling followed by urethral plate fusion in the midline to form urethral seam was observed from ED 16 onwards. Coexpression of epithelial and mesenchymal markers was observed in several cells at the urethral seam; a few cells with coexpression of epithelial and apoptotic markers were also observed. Mesenchymal transformation of epithelial cells and apoptotic epithelial cells was observed under transmission electron microscope.ConclusionUrethral formation occurs by tubulogenesis, which initiates proximally and progresses distally. This is the first study to demonstrate epithelial-mesenchymal transformation and epithelial cell apoptosis in the urethral seam cells of fetal rats. These findings provide new insights into the mechanisms involved in embryonal development of the urethra.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xing Liu
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Fangyuan Huang
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yang Liu
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xining Cao
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lianju Shen
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chunlan Long
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Dawei He
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tao Lin
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Guanghui Wei
- Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
36
|
Infante CR, Rasys AM, Menke DB. Appendages and gene regulatory networks: Lessons from the limbless. Genesis 2017; 56. [PMID: 29076617 DOI: 10.1002/dvg.23078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 01/19/2023]
Abstract
Among squamate reptiles, dozens of lineages have independently evolved complete or partial limb reduction. This remarkable convergence of limbless and limb-reduced phenotypes provides multiple natural replicates of different ages to explore the evolution and development of the vertebrate limb and the gene regulatory network that controls its formation. The most successful and best known of the limb-reduced squamates are snakes, which evolved a limb-reduced body form more than 100 million years ago. Recent studies have revealed the unexpected finding that many ancient limb enhancers are conserved in the genomes of snakes. Analyses in limbed animals show that many of these limb enhancers are also active during development of the phallus, suggesting that these enhancers may have been retained in snakes due their importance in regulating transcription in the external genitalia. This hypothesis is substantiated by functional tests of snake enhancers, which demonstrate that snake enhancer elements have lost limb function while retaining genital enhancer function. The large degree of overlap in the gene regulatory networks deployed during limb and phallus development may act to constrain the divergence of shared gene network components and the evolution of appendage morphology. Future studies will reveal whether limb regulatory elements have undergone similar functional changes in other lineages of limb-reduced squamates.
Collapse
Affiliation(s)
- Carlos R Infante
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, 85721
| | - Ashley M Rasys
- Department of Cellular Biology, University of Georgia, Athens, Georgia, 30602.,Department of Genetics, University of Georgia, Athens, Georgia, 30602
| | - Douglas B Menke
- Department of Genetics, University of Georgia, Athens, Georgia, 30602
| |
Collapse
|
37
|
Akbari P, Fathollahi A, Mo R, Kavran M, Episalla N, Hui CC, Farhat WA, Hijaz AK. A genetic female mouse model with congenital genitourinary anomalies and adult stages of urinary incontinence. Neurourol Urodyn 2017; 36:1981-1987. [PMID: 28244147 DOI: 10.1002/nau.23230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/28/2016] [Accepted: 01/10/2017] [Indexed: 11/10/2022]
Abstract
AIMS To characterize the urinary incontinence observed in adult Gli2+/- ; Gli3Δ699/+ female mice and identify the defects underlying the condition. METHODS Gli2+/- and Gli3Δ699/+ mice were crossed to generate: wild-type, mutant Gli2 (Gli2+/- ), mutant Gli3 (Gli3Δ699/+ ), and double mutant (Gli2+/- ; Gli3Δ699/+ ) female mice, verified via Polymerase Chain Reactions. Bladder functional studies including cystometrogram (CMG), leak point pressure (LPP), and voiding testing were performed on adult female mice. Female bladders and urethras were also analyzed via ink injection and histological assays. RESULTS CMG tracing showed no signal corresponding to the filling of the Gli2+/- ; Gli3Δ699/+ bladders. LPP were significantly reduced in Gli2+/- ; Gli3Δ699/+ mice compared to wild-type mice. CMG studies revealed a decrease in peak micturition pressure values in Gli2+/- ; Gli3Δ699/+ mice compared with all other groups. No significant differences between mutant and wild-type mice were detected in urinary output. Histological analyses revealed Gli2+/- ; Gli3Δ699/+ mice exhibited a widened urethra and a decrease in smooth muscle layer thickness in the bladder outlet and urethra, with increased mucosal folding. CONCLUSIONS Gli2+/- ; Gli3Δ699/+ adult female mice display persistent urinary incontinence due to the malformation of the bladder outlet and urethra. This presents a consistent and reliable genetic mouse model for female urinary incontinence and alludes to the key role of genetic factors involved in the condition.
Collapse
Affiliation(s)
- Pedram Akbari
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Rong Mo
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael Kavran
- Department of Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Nicole Episalla
- Georgetown University School of Medicine, Washington, DC, Washington
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Walid A Farhat
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada.,Division of Urology, Department of Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Adonis K Hijaz
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, Ohio
| |
Collapse
|
38
|
ISL1 is a major susceptibility gene for classic bladder exstrophy and a regulator of urinary tract development. Sci Rep 2017; 7:42170. [PMID: 28176844 PMCID: PMC5296905 DOI: 10.1038/srep42170] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/06/2017] [Indexed: 01/05/2023] Open
Abstract
Previously genome-wide association methods in patients with classic bladder exstrophy (CBE) found association with ISL1, a master control gene expressed in pericloacal mesenchyme. This study sought to further explore the genetics in a larger set of patients following-up on the most promising genomic regions previously reported. Genotypes of 12 markers obtained from 268 CBE patients of Australian, British, German Italian, Spanish and Swedish origin and 1,354 ethnically matched controls and from 92 CBE case-parent trios from North America were analysed. Only marker rs6874700 at the ISL1 locus showed association (p = 2.22 × 10−08). A meta-analysis of rs6874700 of our previous and present study showed a p value of 9.2 × 10−19. Developmental biology models were used to clarify the location of ISL1 activity in the forming urinary tract. Genetic lineage analysis of Isl1-expressing cells by the lineage tracer mouse model showed Isl1-expressing cells in the urinary tract of mouse embryos at E10.5 and distributed in the bladder at E15.5. Expression of isl1 in zebrafish larvae staged 48 hpf was detected in a small region of the developing pronephros. Our study supports ISL1 as a major susceptibility gene for CBE and as a regulator of urinary tract development.
Collapse
|
39
|
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
|
40
|
He F, Akbari P, Mo R, Zhang JJ, Hui CC, Kim PC, Farhat WA. Adult Gli2+/-;Gli3Δ699/+ Male and Female Mice Display a Spectrum of Genital Malformation. PLoS One 2016; 11:e0165958. [PMID: 27814383 PMCID: PMC5096680 DOI: 10.1371/journal.pone.0165958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/20/2016] [Indexed: 11/23/2022] Open
Abstract
Disorders of sexual development (DSD) encompass a broad spectrum of urogenital malformations and are amongst the most common congenital birth defects. Although key genetic factors such as the hedgehog (Hh) family have been identified, a unifying postnatally viable model displaying the spectrum of male and female urogenital malformations has not yet been reported. Since human cases are diagnosed and treated at various stages postnatally, equivalent mouse models enabling analysis at similar stages are of significant interest. Additionally, all non-Hh based genetic models investigating DSD display normal females, leaving female urogenital development largely unknown. Here, we generated compound mutant mice, Gli2+/–;Gli3Δ699/+, which exhibit a spectrum of urogenital malformations in both males and females upon birth, and also carried them well into adulthood. Analysis of embryonic day (E)18.5 and adult mice revealed shortened anogenital distance (AGD), open ventral urethral groove, incomplete fusion of scrotal sac, abnormal penile size and structure, and incomplete testicular descent with hypoplasia in male mice, whereas female mutant mice displayed reduced AGD, urinary incontinence, and a number of uterine anomalies such as vaginal duplication. Male and female fertility was also investigated via breeding cages, and it was identified that male mice were infertile while females were unable to deliver despite becoming impregnated. We propose that Gli2+/–;Gli3Δ699/+ mice can serve as a genetic mouse model for common DSD such as cryptorchidism, hypospadias, and incomplete fusion of the scrotal sac in males, and a spectrum of uterine and vaginal abnormalities along with urinary incontinence in females, which could prove essential in revealing new insights into their equivalent diseases in humans.
Collapse
Affiliation(s)
- Fei He
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Pedram Akbari
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rong Mo
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jennifer J. Zhang
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Peter C. Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, DC, United States of America
| | - Walid A. Farhat
- Program in Developmental & Stem Cell Biology, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Urology, Department of Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
41
|
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
|
42
|
Gredler ML. Developmental and Evolutionary Origins of the Amniote Phallus. Integr Comp Biol 2016; 56:694-704. [DOI: 10.1093/icb/icw102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
43
|
Frankel TE, Meyer MT, Orlando EF. Aqueous exposure to the progestin, levonorgestrel, alters anal fin development and reproductive behavior in the eastern mosquitofish (Gambusia holbrooki). Gen Comp Endocrinol 2016; 234:161-9. [PMID: 26795917 DOI: 10.1016/j.ygcen.2016.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 01/18/2023]
Abstract
Endogenous progestogens are important regulators of vertebrate reproduction. Synthetic progestins are components of human contraceptive and hormone replacement pharmaceuticals. Both progestogens and progestins enter the environment through a number of sources, and have been shown to cause profound effects on reproductive health in various aquatic vertebrates. Progestins are designed to bind human progesterone receptors, but they also have been shown to strongly activate androgen receptors in fish. Levonorgestrel (LNG) activates fish androgen receptors and induces development of male secondary sex characteristics in females of other species. Although behavior has been postulated to be a sensitive early indicator of exposure to certain environmental contaminants, no such research on the reproductive behavior of gestagen-exposed fish has been conducted to date. The goal of our study was to examine the exposure effects of a human contraceptive progestin, LNG, on the reproductive development and behavior of the viviparous eastern mosquitofish (Gambusia holbrooki). Internal fertilization is a requisite characteristic of viviparous species, and is enabled by an androgen driven elongation of the anal fin into the male gonopodium (i.e., phallus). In this study, we exposed adult mosquitofish to ethanol (EtOH control), 10ng/L, and 100ng/L LNG for 8d using a static replacement exposure design. After 8d, a subset of males and females from each treatment were examined for differences in the 4:6 anal fin ratio. In addition, paired social interaction trials were performed using individual control males and control females or females treated 10ng/L or 100ng/L LNG. Female mosquitofish exposed to LNG were masculinized as evidenced by the elongation of the anal fin rays, a feature normal to males and abnormal to females. LNG caused significant increases in the 4:6 anal fin ratios of female mosquitofish in both the 10ng/L and 100ng/L treatments, although these differences were not significant between the two treatments. LNG caused significant increases in the 4:6 anal fin ratio of males exposed to 100ng/L, with no effects observed in the 10ng/L treatment. In addition, the reproductive behavior of control males paired with female mosquitofish exposed to 100ng/L LNG was also altered, for these males spent more time exhibiting no reproductive behavior, had decreased attending behavior, and a lower number of gonopodial thrusts compared to control males paired to control female mosquitofish. Given the rapid effects on both anal fin morphology and behavior observed in this study, the mosquitofish is an excellent sentinel species for the detection of exposure to LNG and likely other 19-nortestosterone derived contraceptive progestins in the environment.
Collapse
Affiliation(s)
- Tyler E Frankel
- University of Maryland, Department of Animal and Avian Sciences, College Park 20742, USA.
| | - Michael T Meyer
- U.S. Geological Survey, Organic Geochemistry Research Laboratory, 4821 Quail Crest Place, Lawrence, KS 66049, USA.
| | - Edward F Orlando
- University of Maryland, Department of Animal and Avian Sciences, College Park 20742, USA
| |
Collapse
|
44
|
Friedman AA, Zelkovic PF, Reda EF, Franco I, Palmer LS. Male and female aphallia associated with severe urinary tract dysplasia. J Pediatr Urol 2016; 12:268.e1-7. [PMID: 27522318 DOI: 10.1016/j.jpurol.2016.04.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/24/2016] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Aphallia is exceedingly rare (1/30 million births). Previous reports have provided limited detail on associated urinary tract findings. OBJECTIVE We reviewed urinary tract anomalies in two boys with aphallia (patients 1 and 2) and a girl with urinary tract dysplasia, a similar external appearance and lack of corporal tissue (patient 3), also consistent with aphallia. CASE REPORTS (FIGURE) Patients 1 and 2 both had a 46XY karyotype, bilateral descended testes in well-formed scrotums, and posterior skin tags containing rudimentary urethras. Patient 1 had a focal area of urethral narrowing; a posterior bladder diverticulum, which drained a ureter; bilateral grade 5 vesicoureteral reflux, with a right partial renal duplication; and hydronephrosis of all moieties. Patient 2 had posterior urethral valves and a bladder diverticulum. Right ureterovesical junction obstruction required a tapered reimplant and later conversion to right-to-left transureteroureterostomy. Patient 3 had a 46XX karyotype and fused, well-formed labia majora. A posterior skin tag was associated with a stenotic urogenital sinus, beyond which were a vagina posteriorly and a right refluxing ureter anteriorly. The left ureter was absent, and a miniscule pouch represented a maldeveloped or absent bladder. Laparoscopy revealed ovaries and normal Müllerian structures. Bilateral renal dysplasia necessitated renal transplant and the creation of an ileocecal neobladder and Mitrofanoff channel. Corporal tissue was diminutive or absent in all. DISCUSSION We see from these three patients that corporal tissue absence can occur in both male and female patients. We propose that the term aphallia can apply to both sexes, as it is the absence of corporal tissue that defines this condition. This is the only report to include and characterize findings in both male and female aphallia patients. Labioscrotal folds develop with a smooth appearance, and, posteriorly, a urethral orifice or Urogenital (UG) sinus with skin tag may be seen. Obstruction at the level of the urethra was common. Severe urinary tract dysplasia was seen in all, a finding not consistently seen or characterized in previous reports. CONCLUSION In girls with severe urinary tract dysplasia and characteristic genital ambiguity, aphallia should be considered. Co-occurrence of aphallia and severe urinary tract dysplasia warrants further urinary tract imaging in all aphallia patients, including voiding cystourethrography, renal bladder ultrasound, and serum creatinine level. Urinary tract reconstruction may be performed without hampering future penile reconstruction, due to modern phallic reconstructive techniques.
Collapse
Affiliation(s)
- Ariella A Friedman
- Cohen Children's Medical Center, Hofstra Northwell School of Medicine, New Hyde Park, NY, USA; Maria Fareri Children's Hospital, Westchester Medical Center, Valhalla, NY, USA.
| | - Paul F Zelkovic
- Maria Fareri Children's Hospital, Westchester Medical Center, Valhalla, NY, USA
| | - Edward F Reda
- Cohen Children's Medical Center, Hofstra Northwell School of Medicine, New Hyde Park, NY, USA; Maria Fareri Children's Hospital, Westchester Medical Center, Valhalla, NY, USA
| | - Israel Franco
- Maria Fareri Children's Hospital, Westchester Medical Center, Valhalla, NY, USA
| | - Lane S Palmer
- Cohen Children's Medical Center, Hofstra Northwell School of Medicine, New Hyde Park, NY, USA
| |
Collapse
|
45
|
Poulet FM, Veneziale R, Vancutsem PM, Losco P, Treinen K, Morrissey RE. Ziracin-Induced Congenital Urogenital Malformations in Female Rats. Toxicol Pathol 2016; 33:320-8. [PMID: 15814361 DOI: 10.1080/01926230590925061] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Spontaneous hypospadias is seldom observed in rats in contrast to its occurrence in 1 out of 250 human births. Ziracin, an antibacterial of the everninomycin class under development for serious enterococcal, staphylococcal, and streptococcal infections, caused anomalies of the external genitalia in F1 female rats and decreased reproductive performance. To characterize the urogenital malformations and determine the period of sensitivity to the effects of Ziracin during development, pregnant rats (F0) were administered 60 mg/kg IV of Ziracin from GD6 to LD21, GD6 to 13, GD14 to the last day of gestation or LD0 to 21. Controls received saline or placebo from GD6 to LD21. Ziracin-induced changes occurred in F1 rats exposed from GD6 to LD21 and GD14 to the last day of gestation, indicating that the period of sensitivity to Ziracin was from GD 14 to the last day of gestation. The urogenital abnormalities consisted of cranial displacement of the urethral opening within the vagina from its normal location at the tip of the genital tubercle. When the urethrovaginal junction occurred at the distal third of the vagina, it created an urogenital cloaca. As a result, ascending infections were seen in the urinary and genital tract. No differences in survivability, body weight, and date of vaginal opening were observed in F1 females. The estrous cycles were slightly prolonged. The mating and fertility indices were decreased as a result of the urogenital anomalies. The mammary glands of pregnant F1 females were underdeveloped, thus F2 pups from affected F1 females had a decreased survival rate. Although the cause of these effects is not known, the findings are consistent with a potential hormonal mechanism.
Collapse
|
46
|
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
|
47
|
Computational modeling and simulation of genital tubercle development. Reprod Toxicol 2016; 64:151-61. [PMID: 27180093 DOI: 10.1016/j.reprotox.2016.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/13/2016] [Accepted: 05/07/2016] [Indexed: 11/22/2022]
Abstract
Hypospadias is a developmental defect of urethral tube closure that has a complex etiology involving genetic and environmental factors, including anti-androgenic and estrogenic disrupting chemicals; however, little is known about the morphoregulatory consequences of androgen/estrogen balance during genital tubercle (GT) development. Computer models that predictively model sexual dimorphism of the GT may provide a useful resource to translate chemical-target bipartite networks and their developmental consequences across the human-relevant chemical universe. Here, we describe a multicellular agent-based model of genital tubercle (GT) development that simulates urethrogenesis from the sexually-indifferent urethral plate stage to urethral tube closure. The prototype model, constructed in CompuCell3D, recapitulates key aspects of GT morphogenesis controlled by SHH, FGF10, and androgen pathways through modulation of stochastic cell behaviors, including differential adhesion, motility, proliferation, and apoptosis. Proper urethral tube closure in the model was shown to depend quantitatively on SHH- and FGF10-induced effects on mesenchymal proliferation and epithelial apoptosis-both ultimately linked to androgen signaling. In the absence of androgen, GT development was feminized and with partial androgen deficiency, the model resolved with incomplete urethral tube closure, thereby providing an in silico platform for probabilistic prediction of hypospadias risk across combinations of minor perturbations to the GT system at various stages of embryonic development.
Collapse
|
48
|
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
|
49
|
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
|
50
|
Jiang JT, Zhong C, Zhu YP, Xu DL, Wood K, Sun WL, Li EH, Liu ZH, Zhao W, Ruan Y, Xia SJ. Prenatal exposure to di-n-butyl phthalate (DBP) differentially alters androgen cascade in undeformed versus hypospadiac male rat offspring. Reprod Toxicol 2016; 61:75-81. [PMID: 26948521 DOI: 10.1016/j.reprotox.2016.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/20/2016] [Accepted: 02/23/2016] [Indexed: 12/01/2022]
Abstract
This study was to compare the alterations of androgen cascades in di-n-butyl phthalate (DBP)-exposed male offspring without hypospadias (undeformed) versus those with hypospadias. To induce hypospadias in male offspring, pregnant rats received DBP via oral gavage at a dose of 750mg/kg BW/day during gestational days 14-18. The mRNA expression levels of genes downstream of the androgen signaling pathway, such as androgen receptor (AR) and Srd5a2, in testes of undeformed rat pups were similar to those in controls; in hypospadiac rat pups these levels were significantly lower than those of control pups. In contrast, both undeformed and hypospadiac rats had decreased serum testosterone levels, reduced mRNA expression of key enzymes in the androgen synthetic pathway in the testes, and ablated genes of developmental pathways, such as Shh, Bmp4, Fgf8, Fgf10 and Fgfr2, in the genital tubercle (GT) as compared to those in DBP-unexposed controls, albeit hypospadiac rats had a more severe decrement than those of undeformed rats. Although other possibilities cannot be excluded, our findings suggest that the relatively normal levels of testosterone-AR-Srd5a2 may contribute to the resistance to DBP toxicity in undeformed rats. In conclusion, our results showed a potential correlation between decreased testosterone levels, reduced mRNA expression of AR and Srd5a2 and the occurrence of hypospadias in male rat offspring prenatally exposed to DBP.
Collapse
Affiliation(s)
- Jun-Tao Jiang
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Chen Zhong
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Yi-Ping Zhu
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Dong-Liang Xu
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Kristofer Wood
- Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W Thomas Rd, Phoenix, AZ 85013, United States
| | - Wen-Lan Sun
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - En-Hui Li
- Department of Urology, Zhejiang Provincial People's Hospital, 158 Shangtang Road, Hangzhou 310014, China
| | - Zhi-Hong Liu
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Wei Zhao
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Yuan Ruan
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China
| | - Shu-Jie Xia
- Department of Urology, Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai 200080, China.
| |
Collapse
|