1
|
Zhang B, Hu P, Wu X, Zheng L, Li X, Wang K, Han X, Wang Y, Hong Y, Qiao R. Mining of candidate genes related to body size in Chinese native pig breeds based on public data. Sci Rep 2025; 15:9793. [PMID: 40118904 PMCID: PMC11928613 DOI: 10.1038/s41598-025-88583-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/29/2025] [Indexed: 03/24/2025] Open
Abstract
To study the key genes that influence the body size of local pig breeds in China. Genome-wide SNP chip data from a total of 129 pigs from eight breeds, consisting of four large body size breeds (MZ, HT, ST, RC) and four small body size breeds (XI, BX, WZ, DN) were analyzed. Principal Component Analysis (PCA) was employed to assess the genetic clustering of the eight breeds. Fst and XP-CLR were used to detect selective signals between the large ans small body size breeds groups. The PCA results indicated a clear clustering of small breeds and a dispersion distribution among large breeds. Fst and XP-CLR identified 142 overlapping regions within a 500 kb up & down stream of significant loci. These regions encompassed 520 annotated genes, which were enriched in 34 biological pathways. Gene network analysis highlighted nine key genes, of which five (NPR3, TNFSF11, TBC1D7, FGF2, IGF1R) are known to be associated with bone growth and body size traits in animals. Additionally, four novel candidate genes (IKBKB, SFRP1, LRP6, SPRY1) were identified that might be related to pig body size. Our findings provide a theoretical basis for further revealing the genetic mechanism of pig body size traits.
Collapse
Affiliation(s)
- Ben Zhang
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Panyang Hu
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiangzhe Wu
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Lixiang Zheng
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiuling Li
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Kejun Wang
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xuelei Han
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yining Wang
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yuan Hong
- College of Animal Science and Technology, FuJian Vocational College of Agriculture, FuZhou, 350119, China.
| | - Ruimin Qiao
- College of Animal Science, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
2
|
Tooze RS, Calpena E, Twigg SRF, D'Arco F, Wakeling EL, Wilkie AOM. Craniosynostosis, inner ear, and renal anomalies in a child with complete loss of SPRY1 (sprouty homolog 1) function. J Med Genet 2023; 60:712-716. [PMID: 36543535 PMCID: PMC10359576 DOI: 10.1136/jmg-2022-108946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION SPRY1 encodes protein sprouty homolog 1 (Spry-1), a negative regulator of receptor tyrosine kinase signalling. Null mutant mice display kidney/urinary tract abnormalities and altered size of the skull; complete loss-of-function of Spry-1 in humans has not been reported. METHODS Analysis of whole-genome sequencing data from individuals with craniosynostosis enrolled in the 100,000 Genomes Project identified a likely pathogenic variant within SPRY1. Reverse-transcriptase PCR and western blot analysis were used to investigate the effect of the variant on SPRY1 mRNA and protein, in lymphoblastoid cell lines from the patient and both parents. RESULTS A nonsense variant in SPRY1, encoding p.(Leu27*), was confirmed to be heterozygous in the unaffected parents and homozygous in the child. The child's phenotype, which included sagittal craniosynostosis, subcutaneous cystic lesions overlying the lambdoid sutures, hearing loss associated with bilateral cochlear and vestibular dysplasia and a unilateral renal cyst, overlapped the features reported in Spry1-/- null mice. Functional studies supported escape from nonsense-mediated decay, but western blot analysis demonstrated complete absence of full-length protein in the affected child and a marked reduction in both parents. CONCLUSION This is the first report of complete loss of Spry-1 function in humans, associated with abnormalities of the cranial sutures, inner ear, and kidneys.
Collapse
Affiliation(s)
- Rebecca S Tooze
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Eduardo Calpena
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Stephen R F Twigg
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Felice D'Arco
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Emma L Wakeling
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Andrew O M Wilkie
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Ebenezer Samuel King JP, Sinha MK, Kumaresan A, Nag P, Das Gupta M, Arul Prakash M, Talluri TR, Datta TK. Cryopreservation process alters the expression of genes involved in pathways associated with the fertility of bull spermatozoa. Front Genet 2022; 13:1025004. [PMID: 36386822 PMCID: PMC9640914 DOI: 10.3389/fgene.2022.1025004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/03/2022] [Indexed: 08/22/2023] Open
Abstract
In bovines, cryopreserved semen is used for artificial insemination; however, the fertility of cryopreserved semen is far lower than that of fresh semen. Although cryopreservation alters sperm phenotypic characteristics, its effect on sperm molecular health is not thoroughly understood. The present study applied next-generation sequencing to investigate the effect of cryopreservation on the sperm transcriptomic composition of bull spermatozoa. While freshly ejaculated bull spermatozoa showed 14,280 transcripts, cryopreserved spermatozoa showed only 12,375 transcripts. Comparative analysis revealed that 241 genes were upregulated, 662 genes were downregulated, and 215 genes showed neutral expression in cryopreserved spermatozoa compared to fresh spermatozoa. Gene ontology analysis indicated that the dysregulated transcripts were involved in nucleic acid binding, transcription-specific activity, and protein kinase binding involving protein autophosphorylation, ventricular septum morphogenesis, and organ development. Moreover, the dysregulated genes in cryopreserved spermatozoa were involved in pathways associated with glycogen metabolism, MAPK signalling, embryonic organ morphogenesis, ectodermal placode formation, and regulation of protein auto-phosphorylation. These findings suggest that the cryopreservation process induced alterations in the abundance of sperm transcripts related to potential fertility-associated functions and pathways, which might partly explain the reduced fertility observed with cryopreserved bull spermatozoa.
Collapse
Affiliation(s)
- John Peter Ebenezer Samuel King
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Manish Kumar Sinha
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Arumugam Kumaresan
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Pradeep Nag
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Mohua Das Gupta
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Mani Arul Prakash
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | - Thirumala Rao Talluri
- Theriogenology Laboratory, Veterinary Gynaecology and Obstetrics, Southern Regional Station of ICAR-National Dairy Research Institute, Bengaluru, Karnataka
| | | |
Collapse
|
4
|
Altés G, Vaquero M, Cuesta S, Anerillas C, Macià A, Espinet C, Ribera J, Bellusci S, Klein OD, Yeramian A, Dolcet X, Egea J, Encinas M. A dominant negative mutation uncovers cooperative control of caudal Wolffian duct development by Sprouty genes. Cell Mol Life Sci 2022; 79:514. [PMID: 36098804 PMCID: PMC9470706 DOI: 10.1007/s00018-022-04546-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022]
Abstract
The Wolffian ducts (WD) are paired epithelial tubules central to the development of the mammalian genitourinary tract. Outgrowths from the WD known as the ureteric buds (UB) generate the collecting ducts of the kidney. Later during development, the caudal portion of the WD will form the vas deferens, epididymis and seminal vesicle in males, and will degenerate in females. While the genetic pathways controlling the development of the UB are firmly established, less is known about those governing development of WD portions caudal to the UB. Sprouty proteins are inhibitors of receptor tyrosine kinase (RTK) signaling in vivo. We have recently shown that homozygous mutation of a conserved tyrosine (Tyr53) of Spry1 results in UB defects indistinguishable from that of Spry1 null mice. Here, we show that heterozygosity for the Spry1 Y53A allele causes caudal WD developmental defects consisting of ectopically branched seminal vesicles in males and persistent WD in females, without affecting kidney development. Detailed analysis reveals that this phenotype also occurs in Spry1+/– mice but with a much lower penetrance, indicating that removal of tyrosine 53 generates a dominant negative mutation in vivo. Supporting this notion, concomitant deletion of one allele of Spry1 and Spry2 also recapitulates the genital phenotype of Spry1Y53A/+ mice with high penetrance. Mechanistically, we show that unlike the effects of Spry1 in kidney development, these caudal WD defects are independent of Ret signaling, but can be completely rescued by lowering the genetic dosage of Fgf10. In conclusion, mutation of tyrosine 53 of Spry1 generates a dominant negative allele that uncovers fine-tuning of caudal WD development by Sprouty genes.
Collapse
Affiliation(s)
- Gisela Altés
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Marta Vaquero
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Sara Cuesta
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain.,Fundación de Investigación Biomédica de Cádiz, Hospital Universitario Puerta del Mar, Novena Planta, Investigación, Av Ana de Viya, 21, 11009, Cádiz, Spain
| | - Carlos Anerillas
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Anna Macià
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Carme Espinet
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Joan Ribera
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | | | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, USA.,Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, USA
| | - Andree Yeramian
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Xavi Dolcet
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Joaquim Egea
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Mario Encinas
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
5
|
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
|
6
|
Chang J, Wang S, Zheng Z. Etiology of Hypospadias: A Comparative Review of Genetic Factors and Developmental Processes Between Human and Animal Models. Res Rep Urol 2021; 12:673-686. [PMID: 33381468 PMCID: PMC7769141 DOI: 10.2147/rru.s276141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/28/2020] [Indexed: 11/23/2022] Open
Abstract
Hypospadias is a congenital anomaly of the penis with an occurrence of approximately 1 in 200 boys, but the etiology of the majority of hypospadias has remained unknown. Numerous genes have been reported as having variants in hypospadias patients, and many studies on genetic deletion of key genes in mouse genital development have also been published. Until now, no comparative analysis in the genes related literature has been reported. The basic knowledge of penile development and hypospadias is mainly obtained from animal model studies. Understanding of the differences and similarities between human and animal models is crucial for studies of hypospadias. In this review, mutations and polymorphisms of hypospadias-related genes have been compared between humans and mice, and differential genotype–phenotype relationships of certain genes between humans and mice have been discussed using the data available in PubMed and MGI online databases, and our analysis only revealed mutations in seven out of 43 human hypospadias related genes which have been reported to show similar phenotypes in mutant mice. The differences and similarities in the processes of penile development and hypospadias malformation among human and commonly used animal models suggest that the guinea pig may be a good model to study the mechanism of human penile development and etiology of hypospadias.
Collapse
Affiliation(s)
- Jun Chang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA.,School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, People's Republic of China
| | - Shanshan Wang
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Zhengui Zheng
- Department of Physiology, School of Medicine, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| |
Collapse
|
7
|
Huh SH, Ha L, Jang HS. Nephron Progenitor Maintenance Is Controlled through Fibroblast Growth Factors and Sprouty1 Interaction. J Am Soc Nephrol 2020; 31:2559-2572. [PMID: 32753399 DOI: 10.1681/asn.2020040401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/08/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Nephron progenitor cells (NPCs) give rise to all segments of functional nephrons and are of great interest due to their potential as a source for novel treatment strategies for kidney disease. Fibroblast growth factor (FGF) signaling plays pivotal roles in generating and maintaining NPCs during kidney development, but little is known about the molecule(s) regulating FGF signaling during nephron development. Sprouty 1 (SPRY1) is an antagonist of receptor tyrosine kinases. Although SPRY1 antagonizes Ret-GDNF signaling, which modulates renal branching, its role in NPCs is not known. METHODS Spry1, Fgf9, and Fgf20 compound mutant animals were used to evaluate kidney phenotypes in mice to understand whether SPRY1 modulates FGF signaling in NPCs and whether FGF8 functions with FGF9 and FGF20 in maintaining NPCs. RESULTS Loss of one copy of Spry1 counters effects of the loss of Fgf9 and Fgf20, rescuing bilateral renal agenesis premature NPC differentiation, NPC proliferation, and cell death defects. In the absence of SPRY1, FGF9, and FGF20, another FGF ligand, FGF8, promotes nephrogenesis. Deleting both Fgf8 and Fgf20 results in kidney agenesis, defects in NPC proliferation, and cell death. Deleting one copy of Fgf8 reversed the effect of deleting one copy of Spry1, which rescued the renal agenesis due to loss of Fgf9 and Fgf20. CONCLUSIONS SPRY1 expressed in NPCs modulates the activity of FGF signaling and regulates NPC stemness. These findings indicate the importance of the balance between positive and negative signals during NPC maintenance.
Collapse
Affiliation(s)
- Sung-Ho Huh
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska .,Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ligyeom Ha
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
8
|
Quantitative Phosphoproteomics Reveals System-Wide Phosphorylation Network Altered by Spry in Mouse Mammary Stromal Fibroblasts. Int J Mol Sci 2019; 20:ijms20215400. [PMID: 31671542 PMCID: PMC6862705 DOI: 10.3390/ijms20215400] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding the fundamental role of the stroma in normal development and cancer progression has been an emerging focus in recent years. The receptor tyrosine kinase (RTK) signaling pathway has been reported playing critical roles in regulating the normal and cancer microenvironment, but the underlying mechanism is still not very clear. By applying the quantitative phosphoproteomic analysis of Sprouty proteins (SPRYs), generic modulators of RTK signaling and deleted mouse mammary fibroblasts, we quantified a total of 11,215 unique phosphorylation sites. By contrast, 554 phosphorylation sites on 425 proteins had SPRY-responsive perturbations. Of these, 554 phosphosites, 362 sites on 277 proteins, were significantly increased, whereas 192 sites on 167 proteins were decreased. Among the regulated proteins, we identified 31 kinases, 7 phosphatases, and one phosphatase inhibitor that were not systematically characterized before. Furthermore, we reconstructed a phosphorylation network centered on RTK signaling regulated by SPRY. Collectively, this study uncovered a system-wide phosphorylation network regulated by SPRY, providing an additional insight into the complicated RTK signaling pathways involved in the mammary gland microenvironment.
Collapse
|
9
|
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
|
10
|
Morgani SM, Saiz N, Garg V, Raina D, Simon CS, Kang M, Arias AM, Nichols J, Schröter C, Hadjantonakis AK. A Sprouty4 reporter to monitor FGF/ERK signaling activity in ESCs and mice. Dev Biol 2018; 441:104-126. [PMID: 29964027 DOI: 10.1016/j.ydbio.2018.06.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022]
Abstract
The FGF/ERK signaling pathway is highly conserved throughout evolution and plays fundamental roles during embryonic development and in adult organisms. While a plethora of expression data exists for ligands, receptors and pathway regulators, we know little about the spatial organization or dynamics of signaling in individual cells within populations. To this end we developed a transcriptional readout of FGF/ERK activity by targeting a histone H2B-linked Venus fluorophore to the endogenous locus of Spry4, an early pathway target, and generated Spry4H2B-Venus embryonic stem cells (ESCs) and a derivative mouse line. The Spry4H2B-Venus reporter was heterogeneously expressed within ESC cultures and responded to FGF/ERK signaling manipulation. In vivo, the Spry4H2B-Venus reporter recapitulated the expression pattern of Spry4 and localized to sites of known FGF/ERK activity including the inner cell mass of the pre-implantation embryo and the limb buds, somites and isthmus of the post-implantation embryo. Additionally, we observed highly localized reporter expression within adult organs. Genetic and chemical disruption of FGF/ERK signaling, in vivo in pre- and post-implantation embryos, abrogated Venus expression establishing the reporter as an accurate signaling readout. This tool will provide new insights into the dynamics of the FGF/ERK signaling pathway during mammalian development.
Collapse
Affiliation(s)
- Sophie M Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Nestor Saiz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vidur Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Dhruv Raina
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Claire S Simon
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Minjung Kang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | | | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Christian Schröter
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.
| |
Collapse
|
11
|
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
|
12
|
Neben CL, Lo M, Jura N, Klein OD. Feedback regulation of RTK signaling in development. Dev Biol 2017; 447:71-89. [PMID: 29079424 DOI: 10.1016/j.ydbio.2017.10.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
Precise regulation of the amplitude and duration of receptor tyrosine kinase (RTK) signaling is critical for the execution of cellular programs and behaviors. Understanding these control mechanisms has important implications for the field of developmental biology, and in recent years, the question of how augmentation or attenuation of RTK signaling via feedback loops modulates development has become of increasing interest. RTK feedback regulation is also important for human disease research; for example, germline mutations in genes that encode RTK signaling pathway components cause numerous human congenital syndromes, and somatic alterations contribute to the pathogenesis of diseases such as cancers. In this review, we survey regulators of RTK signaling that tune receptor activity and intracellular transduction cascades, with a focus on the roles of these genes in the developing embryo. We detail the diverse inhibitory mechanisms utilized by negative feedback regulators that, when lost or perturbed, lead to aberrant increases in RTK signaling. We also discuss recent biochemical and genetic insights into positive regulators of RTK signaling and how these proteins function in tandem with negative regulators to guide embryonic development.
Collapse
Affiliation(s)
- Cynthia L Neben
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA
| | - Megan Lo
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco 94143, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco 94143, USA.
| |
Collapse
|
13
|
de Graaf P, van der Linde EM, Rosier PFWM, Izeta A, Sievert KD, Bosch JLHR, de Kort LMO. Systematic Review to Compare Urothelium Differentiation with Urethral Epithelium Differentiation in Fetal Development, as a Basis for Tissue Engineering of the Male Urethra. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:257-267. [PMID: 27809709 DOI: 10.1089/ten.teb.2016.0352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-engineered (TE) urethra is desirable in men with urethral disease (stricture or hypospadias) and shortage of local tissue. Although ideally a TE graft would contain urethral epithelium cells, currently, bladder epithelium (urothelium) is widely used, but morphologically different. Understanding the differences and similarities of urothelium and urethral epithelium could help design a protocol for in vitro generation of urethral epithelium to be used in TE grafts for the urethra. PURPOSE To understand the development toward urethral epithelium or urothelium to improve TE of the urethra. METHODS A literature search was done following PRISMA guidelines. Articles describing urethral epithelium and bladder urothelium development in laboratory animals and humans were selected. RESULTS Twenty-nine studies on development of urethral epithelium and 29 studies on development of urothelium were included. Both tissue linings derive from endoderm and although adult urothelium and urethral epithelium are characterized by different gene expression profiles, the signaling pathways underlying their development are similar, including Shh, BMP, Wnt, and FGF. The progenitor of the urothelium and the urethral epithelium is the early fetal urogenital sinus (UGS). The urethral plate and the urothelium are both formed from the p63+ cells of the UGS. Keratin 20 and uroplakins are exclusively expressed in urothelium, not in the urethral epithelium. Further research has to be done on unique markers for the urethral epithelium. CONCLUSION This review has summarized the current knowledge about embryonic development of urothelium versus urethral epithelium and especially focuses on the influencing factors that are potentially specific for the eventual morphological differences of both cell linings, to be a basis for developmental or tissue engineering of urethral tissue.
Collapse
Affiliation(s)
- Petra de Graaf
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands .,2 Regenerative Medicine Center Utrecht , Utrecht, The Netherlands
| | | | - Peter F W M Rosier
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Ander Izeta
- 3 Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, Hospital Universitario Donostia , San Sebastián, Spain .,4 Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra , San Sebastián, Spain
| | | | - J L H Ruud Bosch
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Laetitia M O de Kort
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| |
Collapse
|
14
|
Koledova Z, Zhang X, Streuli C, Clarke RB, Klein OD, Werb Z, Lu P. SPRY1 regulates mammary epithelial morphogenesis by modulating EGFR-dependent stromal paracrine signaling and ECM remodeling. Proc Natl Acad Sci U S A 2016; 113:E5731-40. [PMID: 27621461 PMCID: PMC5047180 DOI: 10.1073/pnas.1611532113] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The role of the local microenvironment in influencing cell behavior is central to both normal development and cancer formation. Here, we show that sprouty 1 (SPRY1) modulates the microenvironment to enable proper mammary branching morphogenesis. This process occurs through negative regulation of epidermal growth factor receptor (EGFR) signaling in mammary stroma. Loss of SPRY1 resulted in up-regulation of EGFR-extracellular signal-regulated kinase (ERK) signaling in response to amphiregulin and transforming growth factor alpha stimulation. Consequently, stromal paracrine signaling and ECM remodeling is augmented, leading to increased epithelial branching in the mutant gland. By contrast, down-regulation of EGFR-ERK signaling due to gain of Sprouty function in the stroma led to stunted epithelial branching. Taken together, our results show that modulation of stromal paracrine signaling and ECM remodeling by SPRY1 regulates mammary epithelial morphogenesis during postnatal development.
Collapse
Affiliation(s)
- Zuzana Koledova
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom; Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno 62500, Czech Republic; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaohong Zhang
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Charles Streuli
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom; Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Robert B Clarke
- Breast Cancer Now Research Unit, Institute of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester M20 4QL, United Kingdom
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, CA 94143; Department of Pediatrics, University of California, San Francisco, CA 94143; Program in Craniofacial Biology, University of California, San Francisco, CA 94143; Institute for Human Genetics, University of California, San Francisco, CA 94143
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, CA 94143
| | - Pengfei Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China;
| |
Collapse
|
15
|
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
|
16
|
Gredler ML, Seifert AW, Cohn MJ. Tissue-specific roles of Fgfr2 in development of the external genitalia. Development 2015; 142:2203-12. [PMID: 26081573 DOI: 10.1242/dev.119891] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Congenital anomalies frequently occur in organs that undergo tubulogenesis. Hypospadias is a urethral tube defect defined by mislocalized, oversized, or multiple openings of the penile urethra. Deletion of Fgfr2 or its ligand Fgf10 results in severe hypospadias in mice, in which the entire urethral plate is open along the ventral side of the penis. In the genital tubercle, the embryonic precursor of the penis and clitoris, Fgfr2 is expressed in two epithelial populations: the endodermally derived urethral epithelium and the ectodermally derived surface epithelium. Here, we investigate the tissue-specific roles of Fgfr2 in external genital development by generating conditional deletions of Fgfr2 in each of these cell types. Conditional deletion of Fgfr2 results in two distinct phenotypes: endodermal Fgfr2 deletion causes mild hypospadias and inhibits maturation of a complex urethral epithelium, whereas loss of ectodermal Fgfr2 results in severe hypospadias and absence of the ventral prepuce. Although these cell type-specific mutants exhibit distinctive genital anomalies, cellular analysis reveals that Fgfr2 regulates epithelial maturation and cell cycle progression in the urethral endoderm and in the surface ectoderm. The unexpected finding that ectodermal deletion of Fgfr2 results in the most severe hypospadias highlights a major role for Fgfr2 in the developing genital surface epithelium, where epithelial maturation is required for maintenance of a closed urethral tube. These results demonstrate that urethral tubulogenesis, prepuce morphogenesis, and sexually dimorphic patterning of the lower urethra are controlled by discrete regions of Fgfr2 activity.
Collapse
Affiliation(s)
- Marissa L Gredler
- Department of Biology, 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 Howard Hughes Medical Institute, Department of Molecular Genetics and Microbiology, University of Florida, PO Box 103610, Gainesville, FL 32611, USA
| |
Collapse
|
17
|
Wnt signaling in testis development: Unnecessary or essential? Gene 2015; 565:155-65. [DOI: 10.1016/j.gene.2015.04.066] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/29/2015] [Accepted: 04/24/2015] [Indexed: 11/24/2022]
|
18
|
Harada M, Omori A, Nakahara C, Nakagata N, Akita K, Yamada G. Tissue-specific roles of FGF signaling in external genitalia development. Dev Dyn 2015; 244:759-73. [DOI: 10.1002/dvdy.24277] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/22/2015] [Accepted: 03/22/2015] [Indexed: 11/11/2022] Open
Affiliation(s)
- Masayo Harada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Akiko Omori
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| | - Chiaki Nakahara
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering; Center for Animal Resources and Development, Kumamoto University; Kumamoto Japan
| | - Keiichi Akita
- Department of Clinical Anatomy; Graduate School of Medical and Dental Sciences; Tokyo Medical and Dental University; Tokyo Japan
| | - Gen Yamada
- Institute of Molecular Embryology and Genetics; Kumamoto University; Kumamoto Japan
- Department of Developmental Genetics; Institute of Advanced Medicine; Wakayama Medical University; Wakayama Japan
| |
Collapse
|
19
|
Mattingly A, Finley JK, Knox SM. Salivary gland development and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:573-90. [PMID: 25970268 DOI: 10.1002/wdev.194] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
Mammalian salivary glands synthesize and secrete saliva via a vast interconnected network of epithelial tubes attached to secretory end units. The extensive morphogenesis required to establish this organ is dependent on interactions between multiple cell types (epithelial, mesenchymal, endothelial, and neuronal) and the engagement of a wide range of signaling pathways. Here we describe critical regulators of salivary gland development and discuss how mutations in these impact human organogenesis. In particular, we explore the genetic contribution of growth factor pathways, nerve-derived factors and extracellular matrix molecules to salivary gland formation in mice and humans.
Collapse
Affiliation(s)
- Aaron Mattingly
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer K Finley
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sarah M Knox
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Georgas KM, Armstrong J, Keast JR, Larkins CE, McHugh KM, Southard-Smith EM, Cohn MJ, Batourina E, Dan H, Schneider K, Buehler DP, Wiese CB, Brennan J, Davies JA, Harding SD, Baldock RA, Little MH, Vezina CM, Mendelsohn C. An illustrated anatomical ontology of the developing mouse lower urogenital tract. Development 2015; 142:1893-908. [PMID: 25968320 DOI: 10.1242/dev.117903] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 04/01/2015] [Indexed: 01/10/2023]
Abstract
Malformation of the urogenital tract represents a considerable paediatric burden, with many defects affecting the lower urinary tract (LUT), genital tubercle and associated structures. Understanding the molecular basis of such defects frequently draws on murine models. However, human anatomical terms do not always superimpose on the mouse, and the lack of accurate and standardised nomenclature is hampering the utility of such animal models. We previously developed an anatomical ontology for the murine urogenital system. Here, we present a comprehensive update of this ontology pertaining to mouse LUT, genital tubercle and associated reproductive structures (E10.5 to adult). Ontology changes were based on recently published insights into the cellular and gross anatomy of these structures, and on new analyses of epithelial cell types present in the pelvic urethra and regions of the bladder. Ontology changes include new structures, tissue layers and cell types within the LUT, external genitalia and lower reproductive structures. Representative illustrations, detailed text descriptions and molecular markers that selectively label muscle, nerves/ganglia and epithelia of the lower urogenital system are also presented. The revised ontology will be an important tool for researchers studying urogenital development/malformation in mouse models and will improve our capacity to appropriately interpret these with respect to the human situation.
Collapse
Affiliation(s)
- Kylie M Georgas
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jane Armstrong
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Janet R Keast
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christine E Larkins
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA
| | - Kirk M McHugh
- Centre for Molecular and Human Genetics, The Research Institute at Nationwide Children's Hospital and Division of Anatomy, The Ohio State University, Columbus, OH 43205/10, USA
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA Department of Biology, Genetics Institute, University of Florida, Gainesville, FL 32610, USA Howard Hughes Medical Institute, University of Florida, Gainesville, FL 32610, USA
| | | | - Hanbin Dan
- Columbia University, Department of Urology, New York, NY 10032, USA
| | - Kerry Schneider
- Columbia University, Department of Urology, New York, NY 10032, USA
| | - Dennis P Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Carrie B Wiese
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jane Brennan
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Jamie A Davies
- Center for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Simon D Harding
- MRC Human Genetics Unit, MRC IGMM, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Richard A Baldock
- MRC Human Genetics Unit, MRC IGMM, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Melissa H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Chad M Vezina
- University of Wisconsin-Madison, School of Veterinary Medicine, Madison, WI 53706, USA
| | - Cathy Mendelsohn
- Columbia University, Department of Urology, New York, NY 10032, USA
| |
Collapse
|
21
|
Abstract
Sprouty proteins are evolutionarily conserved modulators of MAPK/ERK pathway. Through interacting with an increasing number of effectors, mediators, and regulators with ultimate influence on multiple targets within or beyond ERK, Sprouty orchestrates a complex, multilayered regulatory system and mediates a crosstalk among different signaling pathways for a coordinated cellular response. As such, Sprouty has been implicated in various developmental and physiological processes. Evidence shows that ERK is aberrantly activated in malignant conditions. Accordingly, Sprouty deregulation has been reported in different cancer types and shown to impact cancer development, progression, and metastasis. In this article, we have tried to provide an overview of the current knowledge about the Sprouty physiology and its regulatory functions in health, as well as an updated review of the Sprouty status in cancer. Putative implications of Sprouty in cancer biology, their clinical relevance, and their proposed applications are also revisited. As a developing story, however, role of Sprouty in cancer remains to be further elucidated.
Collapse
Affiliation(s)
- Samar Masoumi-Moghaddam
- UNSW Department of Surgery, University of New South Wales, St George Hospital, Kogarah, Sydney, NSW, 2217, Australia,
| | | | | |
Collapse
|
22
|
Masoumi-Moghaddam S, Amini A, Wei AQ, Robertson G, Morris DL. Sprouty 1 predicts prognosis in human epithelial ovarian cancer. Am J Cancer Res 2015; 5:1531-1541. [PMID: 26101716 PMCID: PMC4473329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/05/2015] [Indexed: 06/04/2023] Open
Abstract
Sprouty proteins are evolutionary-conserved modulators of receptor tyrosine kinase (RTK) signaling. We have previously reported inverse correlation of the Sprouty 1 (Spry1) protein expression with ovarian cancer cell proliferation, migration, invasion and survival. In the present study, the expression status of Spry1 protein and its clinical relevance in patients with epithelial ovarian cancer were explored. Matched tumor and normal tissue samples from 100 patients with epithelial ovarian cancer were immunohistochemically stained for Spry1. Expression of ERK, p-ERK, Ki67, FGF-2, VEGF and IL-6 and their correlation with Spry1 were also evaluated. In addition, correlation between Spry1 and clinicopathological characteristics and predictive significance of Spry1 for overall survival (OS) and disease-free survival (DFS) were analysed. Our data indicated that Spry1 was significantly downregulated in tumor tissues (p=0.004). Spry1 showed significant inverse correlation with p-ERK/ERK (p=0.045), Ki67 (p=0.010), disease stage (p=0.029), tumor grade (p=0.037), recurrence (p=0.001) and lymphovascular invasion (p=0.042). It was revealed that Spry1 low-expressing patients had significantly poorer OS (p=0.010) and DFS (p=0.012) than those with high expression of Spry1. Multivariate analysis showed that high Spry1 (p=0.030), low stage (p=0.048) and no residual tumor (p=0.007) were independent prognostic factors for a better OS, among which high Spry1 (p=0.035) and low stage (p=0.035) remained as independent predictors of DFS, too. We also found that the expression of Spry1 significantly correlates with the expression of Spry2 (p<0.001), but not that of Spry4. In conclusion, we report for the first time to our knowledge that Spry1 protein is downregulated in human epithelial ovarian cancer. Spry1 expression significantly impacts tumor behavior and shows predictive value as an independent prognostic factor for survival and recurrence.
Collapse
Affiliation(s)
- Samar Masoumi-Moghaddam
- Department of Surgery, St George Hospital, The University of New South WalesGray Street, Kogarah, Sydney NSW 2217, Australia
| | - Afshin Amini
- Department of Surgery, St George Hospital, The University of New South WalesGray Street, Kogarah, Sydney NSW 2217, Australia
| | - Ai-Qun Wei
- Department of Orthopedic Surgery, St George Hospital, The University of New South WalesGray Street, Kogarah, Sydney NSW 2217, Australia
| | - Gregory Robertson
- Department of Gynaecology Oncology, St George Hospital, The University of New South WalesGray Street, Kogarah, Sydney NSW 2217, Australia
| | - David L Morris
- Department of Surgery, St George Hospital, The University of New South WalesGray Street, Kogarah, Sydney NSW 2217, Australia
| |
Collapse
|
23
|
Liu D, Shen L, Tao Y, Kuang Y, Cai L, Wang D, He M, Tong X, Zhou S, Sun J, Shi C, Wang C, Wu Y. Alterations in gene expression during sexual differentiation in androgen receptor knockout mice induced by environmental endocrine disruptors. Int J Mol Med 2014; 35:399-404. [PMID: 25434310 DOI: 10.3892/ijmm.2014.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 11/13/2014] [Indexed: 11/05/2022] Open
Abstract
In the present study, we aimed to explore the effect of environmental endocrine disruptors (EEDs) on sexual differentiation in androgen receptor (AR)-/-, AR+/- and AR+/+ male mice. By using a Cre-loxP conditional knockout strategy, we generated AR knockout mice. By mating flox-AR female mice with AR-Cre male mice, the offspring male mice which were produced were examined. Mice not subjected to any type of intervention were used as the controls. Furthermore, male mice of different genotypes were selected and further divided into subgroups as follows: the control group, bisphenol A (BPA) group and the dibutyl phthalate [corrected] (DBP) group. The expression of the Wilms tumor 1 (WT1), lutropin/choriogonadotropin receptor (LHR), 17-β-hydroxysteroid dehydrogenase type 3 (17βHSD3) and steroid-5-alpha-reductase, alpha polypeptide 2 (SRD5A2) genes was determined by RT-qPCR and western blot analysis. There was no statistically significant difference in the weight of the mice between the control group and the knockout group (P>0.05). The results revealed that, compared with the control group, in the knockout group, anogenital distance was shortened, and testicular weight and testosterone levels were decreased; estradiol levels were elevated; the differences were statistically significant (P<0.05). In the group of AR+/- male mice exposed to 100 mg/l EEDs, hypospadias was successfully induced, suggesting that EEDs are involved in the embryonic stage of sexual development in male mice. The quantitative detection of WT1, LHR, 17βHSD3 and SRD5A2 gene expression by RT-qPCR and western blot analysis indicated that these genes were significantly downregulated in the mice in the BPA group. In conclusion, exposure to EEDs induces hypospadias in heterozygous and wild-type male mice offspring during sexual differentiation, but has no effect on homozygous offspring. Therefore, EEDs play an important role during the third stage of sexual differentiation.
Collapse
Affiliation(s)
- Dehong Liu
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Liping Shen
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yonglin Tao
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Ying Kuang
- Shanghai Research Center for Model Organisms, Shanghai 201203, P.R. China
| | - Lei Cai
- Shanghai Research Center for Model Organisms, Shanghai 201203, P.R. China
| | - Dan Wang
- School of Life Science, Shanghai University, Shanghai 200444, P.R. China
| | - Meiduo He
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xuebo Tong
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Shuguang Zhou
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jie Sun
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Chenchen Shi
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Chunxiao Wang
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yi Wu
- Department of Pediatric Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
24
|
Murashima A, Kishigami S, Thomson A, Yamada G. Androgens and mammalian male reproductive tract development. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:163-70. [PMID: 24875095 DOI: 10.1016/j.bbagrm.2014.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/28/2014] [Accepted: 05/19/2014] [Indexed: 12/31/2022]
Abstract
One of the main functions of androgen is in the sexually dimorphic development of the male reproductive tissues. During embryogenesis, androgen determines the morphogenesis of male specific organs, such as the epididymis, seminal vesicle, prostate and penis. Despite the critical function of androgens in masculinization, the downstream molecular mechanisms of androgen signaling are poorly understood. Tissue recombination experiments and tissue specific androgen receptor (AR) knockout mouse studies have revealed epithelial or mesenchymal specific androgen-AR signaling functions. These findings also indicate that epithelial-mesenchymal interactions are a key feature of AR specific activity, and paracrine growth factor action may mediate some of the effects of androgens. This review focuses on mouse models showing the interactions of androgen and growth factor pathways that promote the sexual differentiation of reproductive organs. Recent studies investigating context dependent AR target genes are also discussed. This article is part of a Special Issue entitled: Nuclear receptors in animal development.
Collapse
Affiliation(s)
- Aki Murashima
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan
| | - Satoshi Kishigami
- Faculty of Biology-Oriented Science and Technology, Kinki University, Kinokawa 649-6493, Wakayama, Japan
| | - Axel Thomson
- Department of Urology, McGill University Health Centre, 1650 Cedar Av, Montreal, Québec, H3A 1A4, Canada
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera 811-1, Wakayama 641-8509, Wakayama, Japan.
| |
Collapse
|