1
|
Sarel-Gallily R, Gunapala KM, Benvenisty N. Large-scale analysis of loss of chromosome Y in human pluripotent stem cells: Implications for Turner syndrome and ribosomopathies. Stem Cell Reports 2025:102471. [PMID: 40185088 DOI: 10.1016/j.stemcr.2025.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Loss of chromosome Y (LOY) occurs in aging and cancers, but its extent and implications in human embryonic stem cells (hESCs) have not been studied. Here, we analyzed over 2,650 RNA sequencing (RNA-seq) samples from hESCs and their differentiated derivatives to detect LOY. We found that 12% of hESC samples have lost their chromosome Y and identified LOY in all three germ layers. Differential expression analysis revealed that LOY samples showed a decrease in expression of pluripotency markers and in ribosomal protein (RP) genes. Strikingly, significant RP transcription downregulation was observed in most RP genes, although there is only one expressed Y-linked RP gene. We further analyzed RP expression in Turner syndrome and Diamond-Blackfan anemia samples and observed overall downregulation of RP transcription. This broad analysis sheds light on the scope and effects of LOY in hESCs, suggesting a novel dosage-sensitive mechanism regulating RP gene transcription in LOY and autosomal ribosomopathies.
Collapse
Affiliation(s)
- Roni Sarel-Gallily
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Keith M Gunapala
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland.
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
2
|
Xue C, Li XH, Ding HQ, Qian X, Zhang MY, Chen K, Wei ZW, Li Y, Jia JH, Zhang WN. Pyridoxine supplementation before puberty ameliorates MAM-induced cognitive and sensorimotor gating impairments. Metab Brain Dis 2024; 40:71. [PMID: 39699742 DOI: 10.1007/s11011-024-01505-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Schizophrenia is a kind of neurodevelopmental mental disorder in which patients begin to experience changes early in their development, typically manifesting around or after puberty and has a fluctuating course. Environmental disturbances during adolescence may be a risk factor for schizophrenia-like deficits. As a better treatment option, preventive intervention prior to schizophrenia may be more beneficial than direct treatment. More effective stress-relieving interventions during the critical puberty period may prevent schizophrenia-like neuronal changes and the transition to schizophrenia in adulthood. Pyridoxine deficiency alters the function of NMDA (n-methyl-D-aspartic acid) receptors and plays a key role in learning and memory. In this study, we prepared a progeny model of schizophrenia by exposing pregnant rats to methoxymethanol acetate (MAM) on gestational day 17. The offspring rats were injected intraperitoneally with pyridoxine daily from birth to prepuberty PND12-PND21), and behavioral changes in the offspring rats were observed in adulthood. Cannabinoid receptor interacting protein 1 (CNRIP1) and cannabinoid receptor-1 (CB1R), which regulate memory, cognitive and motor activity, were detected in the prefrontal cortex (PFC) and hippocampus of the offspring rats, and the cell proliferation in the hippocampal dentate gyrus (DG) was also observed. The results showed that the MAM rats spent less time the open arm in the elevated plus maze test, decreased discrimination coefficient in novel object recognition test, and decreased prepulse inhibition, while the MAM rats supplemented with pyridoxine in prepuberty did not show any of the above abnormal behavioral changes in adulthood. By examining related proteins in the PFC and hippocampus, we found that only CB1R protein expression was downregulated in the PFC, whereas CNRIP1 expression was not only elevated in the hippocampus, but also significantly increased in pyridoxine- supplemented MAM rats. Meanwhile, pyridoxine supplementation alleviated the reduction of doublecortin (DCX)-positive cells and Ki67-positive cells in MAM rats. These results indicate that prepuberty pyridoxine supplementation has a positive effect on the prevention of cognitive deficits and sensorimotor gating impairment in MAM-induced schizophrenia-like rats, accompanied by changes in the CB1R and CNRIP1 expression in PFC and hippocampus, as well as the regeneration of neurons in the DG region.
Collapse
Affiliation(s)
- Cheng Xue
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
- Department of Clinical Laboratory, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, 213003, PR China
| | - Xiao-Hui Li
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
- Department of Clinical Laboratory, Xiangyang First People's Hospital, Hubei University of Medicine, Xiangyang, 441000, PR China
| | - Hong-Qun Ding
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Xin Qian
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Meng-Yu Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Kai Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Zi-Wei Wei
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Ying Li
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China
| | - Jun-Hai Jia
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China.
| | - Wei-Ning Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, PR China.
| |
Collapse
|
3
|
Punetha M, Saini S, Sharma S, Thakur S, Dahiya P, Mangal M, Kumar R, Kumar D, Yadav PS. CRISPR-Mediated SRY Gene Mutation Increases the Expression of Female Lineage-Specific Gene in Pre-Implantation Buffalo Embryo. Reprod Domest Anim 2024; 59:e14739. [PMID: 39498956 DOI: 10.1111/rda.14739] [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: 08/09/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024]
Abstract
In mammals, sex determination is governed by the SRY gene on the Y chromosome, redirecting gonadal development from forming ovaries to testes. Mutations or alterations in the SRY gene can significantly affect phenotypic changes and lineage-specific markers. This study aims to elucidate the role of the SRY gene in buffalo embryos using CRISPR-Cas9 technology. We designed a crRNA targeting the HMG domain of the SRY gene using the CRISPOR algorithm. Nucleofection of sgRNA-Cas9 RNPs into buffalo fibroblasts confirmed efficient cleavage at the targeted site. Using this validated guide, we investigated the role of the SRY gene in sexual determination by electroporating CRISPR-Cas9-RNPs into single-stage zygotes of buffalo. Genetic changes in the SRY gene were confirmed through sequencing, revealing mosaic blastocysts with multiple alleles and non-mosaic mutants. Mutations in SRY gene increased the expression of female lineage-specific gene Wnt4 whereas decreased the expression of male specific gene SOX9 in blastocysts, suggesting reprogramming towards female sex determination pathways. Our findings provide insights into buffalo sex differentiation mechanisms and potential applications in reproductive strategies for breeding programmes.
Collapse
Affiliation(s)
- Meeti Punetha
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Sheetal Saini
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Surabhi Sharma
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Swati Thakur
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Priya Dahiya
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Manu Mangal
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Rajesh Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| | - P S Yadav
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, India
| |
Collapse
|
4
|
Swift KM, Gary NC, Urbanczyk PJ. On the basis of sex and sleep: the influence of the estrous cycle and sex on sleep-wake behavior. Front Neurosci 2024; 18:1426189. [PMID: 39268035 PMCID: PMC11390649 DOI: 10.3389/fnins.2024.1426189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
The recurrent hormonal fluctuations within reproductive cycles impact sleep-wake behavior in women and in rats and mice used in preclinical models of sleep research. Strides have been made in sleep-related clinical trials to include equal numbers of women; however, the inclusion of female rodents in neuroscience and sleep research is lacking. Female animals are commonly omitted from studies over concerns of the effect of estrus cycle hormones on measured outcomes. This review highlights the estrous cycle's broad effects on sleep-wake behavior: from changes in sleep macroarchitecture to regionally specific alterations in neural oscillations. These changes are largely driven by cycle-dependent ovarian hormonal fluctuations occurring during proestrus and estrus that modulate neural circuits regulating sleep-wake behavior. Removal of estrous cycle influence by ovariectomy ablates characteristic sleep changes. Further, sex differences in sleep are present between gonadally intact females and males. Removal of reproductive hormones via gonadectomy in both sexes mitigates some, but not all sex differences. We examine the extent to which reproductive hormones and sex chromosomes contribute to sex differences in sleep-wake behavior. Finally, this review addresses the limitations in our understanding of the estrous cycle's impact on sleep-wake behavior, gaps in female sleep research that are well studied in males, and the implications that ignoring the estrous cycle has on studies of sleep-related processes.
Collapse
Affiliation(s)
- Kevin M Swift
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Nicholas C Gary
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Phillip J Urbanczyk
- Medical Readiness Systems Biology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
5
|
Wang H, Lan S, Wang L, Zhao J, Jia X, Xu J, Sun G, Liu L, Gong S, Wang N, Shan B, Zhang F, Zhang Z. Expression of circ-PHC3 enhances ovarian cancer progression via regulation of the miR-497-5p/SOX9 pathway. J Ovarian Res 2023; 16:142. [PMID: 37468993 DOI: 10.1186/s13048-023-01170-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 04/25/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Accumulating studies have reported indispensable functions of circular RNAs (circRNA) in tumor progression through regulation of gene expression. However, circRNA expression profiles and functions in human ovarian carcinoma (OC) are yet to be fully established. METHODS In this research, deep sequencing of circRNAs from OC samples and paired adjacent normal tissues was performed to establish expression profiles and circ-PHC3 levels between the groups further compared using RT-qPCR. The effects of ectopic overexpression of miR-497-5p and SOX9 and siRNA-mediated knockdown of circ-PHC3 and an miR-497-5p inhibitor were explored to clarify the regulatory mechanisms underlying circ-PHC3 activity in OC proliferation and metastasis. Information from public databases and the luciferase reporter assay were further utilized to examine the potential correlations among circ-PHC3, miR-497-5p and SOX9. RESULTS Our results showed significant upregulation of circ-PHC3 in both OC cell lines and tissues. In the luciferase reporter assay, downregulation of circ-PHC3 led to suppression of metastasis and proliferation, potentially through targeted effects on the miR-497-5p/SOX9 axis in OC. SOX9 overexpression or miR-497-5p suppression rescued OC cell proliferation and invasion following silencing of circ-PHC3. Moreover, SOX9 inhibition induced restoration of OC cell invasion and proliferation under conditions of overexpression of miR-497-5p. Thus, circ-PHC3 appears to exert effects on cancer stem cell differentiation through regulation of the miR-497-5p/SOX9 axis. CONCLUSION Taken together, our findings suggest that circ-PHC3 enhances OC progression through functioning as an miR-497-5p sponge to promote SOX9 expression, supporting its potential as a promising candidate target for OC therapy.
Collapse
Affiliation(s)
- Hongxia Wang
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Suwei Lan
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Lingxiang Wang
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Jingyun Zhao
- Department of Reproductive Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinzhuan Jia
- Department of Reproductive Medicine, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jie Xu
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
- Department of Gynecology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangyu Sun
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Leilei Liu
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Shan Gong
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Na Wang
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China
| | - Baoen Shan
- Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fenghua Zhang
- Department of Breast & Thyroid Surgery, Hebei General Hospital, No.348 Heping West Road, Shijiazhuang, 050051, Hebei, China.
| | - Zhengmao Zhang
- Department of Gynecology, Fourth Hospital of Hebei Medical University, No.12 Jiankang Road, Shijiazhuang, 050011, China.
| |
Collapse
|
6
|
Reyes AP, León NY, Frost ER, Harley VR. Genetic control of typical and atypical sex development. Nat Rev Urol 2023:10.1038/s41585-023-00754-x. [PMID: 37020056 DOI: 10.1038/s41585-023-00754-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 04/07/2023]
Abstract
Sex development relies on the sex-specific action of gene networks to differentiate the bipotential gonads of the growing fetus into testis or ovaries, followed by the differentiation of internal and external genitalia depending on the presence or absence of hormones. Differences in sex development (DSD) arise from congenital alterations during any of these processes, and are classified depending on sex chromosomal constitution as sex chromosome DSD, 46,XY DSD or 46,XX DSD. Understanding the genetics and embryology of typical and atypical sex development is essential for diagnosing, treating and managing DSD. Advances have been made in understanding the genetic causes of DSD over the past 10 years, especially for 46,XY DSD. Additional information is required to better understand ovarian and female development and to identify further genetic causes of 46,XX DSD, besides congenital adrenal hyperplasia. Ongoing research is focused on the discovery of further genes related to typical and atypical sex development and, therefore, on improving diagnosis of DSD.
Collapse
Affiliation(s)
- Alejandra P Reyes
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Genetics Department, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Nayla Y León
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Emily R Frost
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
| | - Vincent R Harley
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Cao Z, Liu L, Bu Z, Yang Z, Li Y, Li R. Bioinformatics analysis and verification of hub genes in 46,XY, disorders of sexual development. Reprod Fertil Dev 2023; 35:353-362. [PMID: 36780715 DOI: 10.1071/rd22134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 01/16/2023] [Indexed: 02/15/2023] Open
Abstract
CONTEXT 46,XY, disorders of sexual development (46,XY, DSD) is a congenital genetic disease whose pathogenesis is complex and clinical manifestations are diverse. The existing molecular research has often focused on single-centre sequencing data, instead of prediction based on big data. AIMS This work aimed to fully understand the pathogenesis of 46,XY, DSD, and summarise the key pathogenic genes. METHODS Firstly, the potential pathogenic genes were identified from public data. Secondly, bioinformatics was used to predict pathogenic genes, including hub gene analysis, protein-protein interaction (PPI) and function enrichment analysis. Lastly, the genomic DNA from two unrelated families were recruited, next-generation sequencing and Sanger sequencing were performed to verify the hub genes. KEY RESULTS A total of 161 potential pathogenic genes were selected from MGI and PubMed gene sets. The PPI network was built which included 144 nodes and 194 edges. MCODE 4 was selected from PPI which scored the most significant P -value. The top 15 hub genes were ranked and identified by Cytoscape. Furthermore, three variants were found on SRD5A2 gene by genome sequencing, which belonged to the prediction hub genes. CONCLUSIONS Our results indicate that occurrence of 46,XY, DSD is attributed to a variety of genes. Bioinformatics analysis can help us predict the hub genes and find the most core network MCODE model. IMPLICATIONS Bioinformatic predictions may provide a novel perspective on better understanding the pathogenesis of 46,XY, DSD.
Collapse
Affiliation(s)
- Zilong Cao
- Ninth Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liqiang Liu
- Ninth Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoyun Bu
- Department of Pediatric Surgery, Rizhao People's Hospital of Shandong Province, Rizhao, Shandong, China
| | - Zhe Yang
- Second Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangqun Li
- Second Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Li
- Ninth Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Ye Z, Bishop T, Wang Y, Shahriari R, Lynch M. Evolution of sex determination in crustaceans. MARINE LIFE SCIENCE & TECHNOLOGY 2023; 5:1-11. [PMID: 37073332 PMCID: PMC10077267 DOI: 10.1007/s42995-023-00163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 12/28/2022] [Indexed: 05/03/2023]
Abstract
Sex determination (SD) involves mechanisms that determine whether an individual will develop into a male, female, or in rare cases, hermaphrodite. Crustaceans harbor extremely diverse SD systems, including hermaphroditism, environmental sex determination (ESD), genetic sex determination (GSD), and cytoplasmic sex determination (e.g., Wolbachia controlled SD systems). Such diversity lays the groundwork for researching the evolution of SD in crustaceans, i.e., transitions among different SD systems. However, most previous research has focused on understanding the mechanism of SD within a single lineage or species, overlooking the transition across different SD systems. To help bridge this gap, we summarize the understanding of SD in various clades of crustaceans, and discuss how different SD systems might evolve from one another. Furthermore, we review the genetic basis for transitions between different SD systems (i.e., Dmrt genes) and propose the microcrustacean Daphnia (clade Branchiopoda) as a model to study the transition from ESD to GSD.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ 85287 USA
| | - Trent Bishop
- Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ 85287 USA
| | - Yaohai Wang
- Institute of Evolution and Marine Biodiversity, KLMME, Ocean University of China, Qingdao, 266003 China
| | - Ryan Shahriari
- Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ 85287 USA
| | - Michael Lynch
- Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ 85287 USA
| |
Collapse
|
9
|
Role of mesonephric contribution to mouse testicular development revisited. Differentiation 2023; 129:109-119. [PMID: 35000816 DOI: 10.1016/j.diff.2021.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 01/25/2023]
Abstract
The role of the mesonephros in testicular development was re-evaluated by growing embryonic day 11.5 (E11.5) mouse testes devoid of mesonephros for 8-21 days in vivo under the renal capsule of castrated male athymic nude mice. This method provides improved growth conditions relative to previous studies based upon short-term (4-7 days) organ culture. Meticulous controls involved wholemount examination of dissected E11.5 mouse testes as well as serial sections of dissected E11.5 mouse testes which were indeed shown to be devoid of mesonephros. As expected, grafts of E11.5 mouse testes with mesonephros attached formed seminiferous tubules and also contained mesonephric derivatives. Grafts of E11.5 mouse testes without associated mesonephros also formed seminiferous tubules and never contained mesonephric derivatives. The consistent absence of mesonephric derivatives in grafts of E11.5 mouse testes grafted alone is further proof of the complete removal of the mesonephros from the E11.5 mouse testes. The testicular tissues that developed in grafts of E11.5 mouse testes alone contained canalized seminiferous tubules composed of Sox9-positive Sertoli cells as well as GENA-positive germ cells. The seminiferous tubules were surrounded by α-actin-positive myoid cells, and the interstitial space contained 3βHSD-1-positive Leydig cells. Grafts of E11.5 GFP mouse testes into wild-type hosts developed GFP-positive vasculature indicating that E11.5 mouse testes contain vascular precursors. These results indicate that the E11.5 mouse testis contains precursor cells for Sertoli cells, Leydig cells, myoid cells and vasculature whose development and differentiation are independent of cells migrating from the E11.5 mesonephros.
Collapse
|
10
|
A model to study human ovotesticular syndrome. Differentiation 2023; 129:60-78. [PMID: 35164980 DOI: 10.1016/j.diff.2021.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/11/2021] [Accepted: 12/12/2021] [Indexed: 01/25/2023]
Abstract
Ovotesticular syndrome is a rare disorder of sex development characterized by the presence of testicular and ovarian tissue. The histologic characteristics of human testicular tissue are well defined by the presence of seminiferous cords or tubules containing TSPY-positive germ cells and Sox9-positive Sertoli cells surrounded by interstitial tissue containing cytochrome P450-positive Leydig cells and smooth muscle α-actin-positive peritubular myoid cells. The histological characteristics of the ovary can be defined by germ cell nests and the development of follicles. In contrast to the testis, the ovary has a paucity of defined specific protein markers, with the granulosa cell marker FOXL2 being the most widely used. In practice, defining the ovarian component of the ovotestis can be quite difficult. We developed a model of human ovotesticular syndrome by combining fetal human testis and ovary in a xenograft model. Ovotesticular xenografts were grown under the renal capsules of gonadectomized athymic nude mice for 6-32 weeks along with age matched control grafts of fetal testis and ovary. Forty ovotesticular xenografts and their controls were analyzed by histology, immunohistochemistry, and fluorescent in situ hybridization to determine the protein expression and karyotype of the cells within the grafts. The ovotesticular xenografts exhibited recognizable testicular and ovarian tissue based on testis-specific and ovary-specific markers defined above. The xenografts simulated a bipolar ovotestis in which the testicular and ovarian elements retain their separate histological characteristics and are separated by a well-defined border. This contrasts with the compartmentalized ovotestis previously described in the literature where the testicular tissue is surrounded by ovarian tissue or a mixed histology where testicular and ovarian tissues are interspersed throughout the gonad. In conclusion, we have characterized a human model of ovotestis which will allow a deeper understanding of ovotestis development in humans and facilitate a more accurate diagnosis of the ovotesticular syndrome.
Collapse
|
11
|
Li Y, Overland M, Derpinghaus A, Aksel S, Cao M, Ladwig N, Cunha GR, Baskin LS. Development of the human fetal testis: Morphology and expression of cellular differentiation markers. Differentiation 2023; 129:17-36. [PMID: 35490077 DOI: 10.1016/j.diff.2022.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 01/25/2023]
Abstract
A comprehensive immunohistochemical ontogeny of the developing human fetal testis has remained incomplete in the literature to date. We collected human fetal testes from 8 to 21 weeks of fetal age, as well as postnatal human testes at minipuberty, pre-pubertal, and pubertal stages. Immunohistochemistry was performed with a comprehensive panel of antigens targeting gonadocytes, Sertoli cells, fetal Leydig cells, peritubular myoid cells, and other hormonal and developmental targets. Testicular cords, precursor structures to seminiferous tubules, developed from 8 to 14 weeks of fetal age, separating the testis into the interstitial and intracordal compartments. Fetal gonadocytes were localized within the testicular cords and evaluated for Testis-Specific Protein Y, Octamer-binding transcription factor 4, Sal-like protein 4, and placental alkaline phosphatase expression. Fetal Sertoli cells were also localized in the testicular cords and evaluated for SRY-box Transcription Factor 9, inhibin, and anti-Mullerian hormone expression. Fetal Leydig cells were present in the interstitium and stained for cytochrome p450c17 and calretinin, while interstitial peritubular myoid cells were examined using smooth muscle α-actin staining. Androgen receptor expression was localized close to the testicular medulla at 8 weeks and then around the testicular cords in the interstitium as they matured in structure. Postnatal staining showed that Testis-Specific Protein Y remained positive of male gonadocytes throughout adulthood. Anti-Mullerian hormone, SRY-box Transcription Factor 9, and Steroidogenic factor 1 are expressed by the postnatal Sertoli cells at all ages examined. Leydig cell markers cytochrome p450c17 and calretinin are expressed during mini-puberty and puberty, but not expressed during the pre-pubertal period. Smooth muscle α-actin and androgen receptor were not expressed during mini-puberty or pre-puberty, but again expressed during the pubertal period. The ontogenic map of the human fetal and postnatal testicular structure and expression patterns described here will serve as a reference for future investigations into normal and abnormal testicular development.
Collapse
Affiliation(s)
- Yi Li
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Maya Overland
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Nicholas Ladwig
- Department of Pathology, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| |
Collapse
|
12
|
Anderson NK, Goodwin SE, Schuppe ER, Dawn A, Preininger D, Mangiamele LA, Fuxjager MJ. Activational vs. organizational effects of sex steroids and their role in the evolution of reproductive behavior: Looking to foot-flagging frogs and beyond. Horm Behav 2022; 146:105248. [PMID: 36054981 DOI: 10.1016/j.yhbeh.2022.105248] [Citation(s) in RCA: 2] [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: 05/25/2022] [Revised: 07/14/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022]
Abstract
Sex steroids play an important role in regulation of the vertebrate reproductive phenotype. This is because sex steroids not only activate sexual behaviors that mediate copulation, courtship, and aggression, but they also help guide the development of neural and muscular systems that underlie these traits. Many biologists have therefore described the effects of sex steroid action on reproductive behavior as both "activational" and "organizational," respectively. Here, we focus on these phenomena from an evolutionary standpoint, highlighting that we know relatively little about the way that organizational effects evolve in the natural world to support the adaptation and diversification of reproductive behavior. We first review the evidence that such effects do in fact evolve to mediate the evolution of sexual behavior. We then introduce an emerging animal model - the foot-flagging frog, Staurois parvus - that will be useful to study how sex hormones shape neuromotor development necessary for sexual displays. The foot flag is nothing more than a waving display that males use to compete for access to female mates, and thus the neural circuits that control its production are likely laid down when limb control systems arise during the developmental transition from tadpole to frog. We provide data that highlights how sex steroids might organize foot-flagging behavior through its putative underlying mechanisms. Overall, we anticipate that future studies of foot-flagging frogs will open a powerful window from which to see how sex steroids influence the neuromotor systems to help germinate circuits that drive signaling behavior. In this way, our aim is to bring attention to the important frontier of endocrinological regulation of evolutionary developmental biology (endo-evo-devo) and its relationship to behavior.
Collapse
Affiliation(s)
- Nigel K Anderson
- Department of Ecology, Evolution, and Organismal Biology, Brown University, Providence, RI, United States of America
| | - Sarah E Goodwin
- Department of Biological Sciences, Smith College, Northampton, MA, United States of America
| | - Eric R Schuppe
- Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA, United States of America
| | - AllexAndrya Dawn
- Department of Biological Sciences, Smith College, Northampton, MA, United States of America
| | - Doris Preininger
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria; Vienna Zoo, Vienna, Austria
| | - Lisa A Mangiamele
- Department of Biological Sciences, Smith College, Northampton, MA, United States of America.
| | - Matthew J Fuxjager
- Department of Ecology, Evolution, and Organismal Biology, Brown University, Providence, RI, United States of America.
| |
Collapse
|
13
|
Li M, Liu W, Li J, Zhang H, Xu J. miR-30c plays diagnostic and prognostic roles and mediates epithelial-mesenchymal transition (EMT) and proliferation of gliomas by affecting Notch1. Sci Rep 2022; 12:16404. [PMID: 36180477 PMCID: PMC9525598 DOI: 10.1038/s41598-022-19326-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/27/2022] [Indexed: 12/02/2022] Open
Abstract
miR-30c functions as a tumor suppressor gene in the majority of tumors, including gliomas. In our study, we discovered that the expression levels of miR-30c in glioma tissues and plasma prior to surgery were lower than those in normal brain tissue following brain injury decompression and in plasma in healthy volunteers. The low expression of miR-30c was closely aligned with the WHO grade, tumor size, PFS, and OS. Additionally, the miR-30c expression level in tumor tissue was positively correlated with the levels in preoperative plasma. In cell biology experiments, miR-30c inhibited EMT and proliferation, migration, and invasion of glioma cells. Analysis of databases of miRNA target genes, real-time quantitative PCR, western blotting, and dual luciferase reporter assays demonstrated that Notch1 is the direct target gene of miR-30c. An inhibitor and shRNA-Notch1 were cotransfected into glioma cells, and it was found that shRNA-Notch1 reduced the enhancement of inhibitors of EMT and proliferation, migration, and invasion of glioma cells. Therefore, we believe that when utilized as a tumor suppressor gene, miR-30c can inhibit EMT and the proliferation, migration, and invasion of glioma cells by directly acting on Notch1 at the posttranscriptional level and that it is a potential diagnostic and prognostic marker.
Collapse
Affiliation(s)
- Mengkao Li
- Department of Neurosurgery, People's Hospital of Longhua, Shenzhen, Guangdong Province, People's Republic of China
| | - Wenzhi Liu
- Department of Radiation Oncology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong Province, People's Republic of China. .,The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, People's Republic of China.
| | - Jian Li
- Department of Neurosurgery, Taian Central Hospital, Taian, Shandong Province, People's Republic of China
| | - Hong Zhang
- Department of Clinical Oncology, Taian Central Hospital, Taian, Shandong Province, People's Republic of China
| | - Jin Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, People's Republic of China.
| |
Collapse
|
14
|
Goodman M, Yacoub R, Getahun D, McCracken CE, Vupputuri S, Lash TL, Roblin D, Contreras R, Cromwell L, Gardner MD, Hoffman T, Hu H, Im TM, Prakash Asrani R, Robinson B, Xie F, Nash R, Zhang Q, Bhai SA, Venkatakrishnan K, Stoller B, Liu Y, Gullickson C, Ahmed M, Rink D, Voss A, Jung HL, Kim J, Lee PA, Sandberg DE. Cohort profile: pathways to care among people with disorders of sex development (DSD). BMJ Open 2022; 12:e063409. [PMID: 36130763 PMCID: PMC9494584 DOI: 10.1136/bmjopen-2022-063409] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PURPOSE The 'DSD Pathways' study was initiated to assess health status and patterns of care among people enrolled in large integrated healthcare systems and diagnosed with conditions comprising the broad category of disorders (differences) of sex development (DSD). The objectives of this communication are to describe methods of cohort ascertainment for two specific DSD conditions-classic congenital adrenal hyperplasia with 46,XX karyotype (46,XX CAH) and complete androgen insensitivity syndrome (CAIS). PARTICIPANTS Using electronic health records we developed an algorithm that combined diagnostic codes, clinical notes, laboratory data and pharmacy records to assign each cohort candidate a 'strength-of-evidence' score supporting the diagnosis of interest. A sample of cohort candidates underwent a review of the full medical record to determine the score cutoffs for final cohort validation. FINDINGS TO DATE Among 5404 classic 46,XX CAH cohort candidates the strength-of-evidence scores ranged between 0 and 10. Based on sample validation, the eligibility cut-off for full review was set at the strength-of-evidence score of ≥7 among children under the age of 8 years and ≥8 among older cohort candidates. The final validation of all cohort candidates who met the cut-off criteria identified 115 persons with classic 46,XX CAH. The strength-of-evidence scores among 648 CAIS cohort candidates ranged from 2 to 10. There were no confirmed CAIS cases among cohort candidates with scores <6. The in-depth medical record review for candidates with scores ≥6 identified 61 confirmed cases of CAIS. FUTURE PLANS As the first cohort of this type, the DSD Pathways study is well-positioned to fill existing knowledge gaps related to management and outcomes in this heterogeneous population. Analyses will examine diagnostic and referral patterns, adherence to care recommendations and physical and mental health morbidities examined through comparisons of DSD and reference populations and analyses of health status across DSD categories.
Collapse
Affiliation(s)
- Michael Goodman
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Rami Yacoub
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Darios Getahun
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
- Health Systems Science, Kaiser Permanente Bernard J Tyson School of Medicine, Pasadena, California, USA
| | - Courtney E McCracken
- Center for Research and Evaluation, Kaiser Permanente Georgia, Atlanta, Georgia, USA
| | - Suma Vupputuri
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, Maryland, USA
| | - Timothy L Lash
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
- Aarhus Universitet, Aarhus, Midtjylland, Denmark
| | - Douglas Roblin
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, Maryland, USA
| | - Richard Contreras
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | - Lee Cromwell
- Center for Research and Evaluation, Kaiser Permanente Georgia, Atlanta, Georgia, USA
| | - Melissa D Gardner
- Susan B Meister Child Health and Evaluation Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Trenton Hoffman
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Haihong Hu
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, Maryland, USA
| | - Theresa M Im
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | | | - Brandi Robinson
- Center for Research and Evaluation, Kaiser Permanente Georgia, Atlanta, Georgia, USA
| | - Fagen Xie
- Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA
| | - Rebecca Nash
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Qi Zhang
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Sadaf A Bhai
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | | | - Bethany Stoller
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Yijun Liu
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | | | - Maaz Ahmed
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - David Rink
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Ava Voss
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Hye-Lee Jung
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Jin Kim
- Epidemiology, Rollins School of Public Health, Atlanta, Georgia, USA
| | - Peter A Lee
- Division of Endocrinology, Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - David E Sandberg
- Susan B Meister Child Health and Evaluation Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Frost ER, Ford EA, Peters AE, Lovell-Badge R, Taylor G, McLaughlin EA, Sutherland JM. A New Understanding, Guided by Single-Cell Sequencing, of the Establishment and Maintenance of the Ovarian Reserve in Mammals. Sex Dev 2022; 17:145-155. [PMID: 36122567 DOI: 10.1159/000526426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Oocytes are a finite and non-renewable resource that are maintained in primordial follicle structures. The ovarian reserve is the totality of primordial follicles, present from birth, within the ovary and its establishment, size, and maintenance dictates the duration of the female reproductive lifespan. Understanding the cellular and molecular dynamics relevant to the establishment and maintenance of the reserve provides the first steps necessary for modulating both individual human and animal reproductive health as well as population dynamics. SUMMARY This review details the key stages of establishment and maintenance of the ovarian reserve, encompassing germ cell nest formation, germ cell nest breakdown, and primordial follicle formation and activation. Furthermore, we spotlight several formative single-cell sequencing studies that have significantly advanced our knowledge of novel molecular regulators of the ovarian reserve, which may improve our ability to modulate female reproductive lifespans. KEY MESSAGES The application of single-cell sequencing to studies of ovarian development in mammals, especially when leveraging genetic and environmental models, offers significant insights into fertility and its regulation. Moreover, comparative studies looking at key stages in the development of the ovarian reserve across species has the potential to impact not just human fertility, but also conservation biology, invasive species management, and agriculture.
Collapse
Affiliation(s)
- Emily R Frost
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, UK
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Emmalee A Ford
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Alexandra E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, UK
| | - Güneş Taylor
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London, UK
| | - Eileen A McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Faculty of Science, Medicine & Health, University of Wollongong, Wollongong, New South Wales, Australia
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
16
|
Role of p38 MAPK Signalling in Testis Development and Male Fertility. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6891897. [PMID: 36092154 PMCID: PMC9453003 DOI: 10.1155/2022/6891897] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/31/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022]
Abstract
The testis is an important male reproductive organ, which ensures reproductive function via the secretion of testosterone and the generation of spermatozoa. Testis development begins in the embryonic period, continues after birth, and generally reaches functional maturation at puberty. The stress-activated kinase, p38 mitogen-activated protein kinase (MAPK), regulates multiple cell processes including proliferation, differentiation, apoptosis, and cellular stress responses. p38 MAPK signalling plays a crucial role in testis development by regulating spermatogenesis, the fate determination of pre-Sertoli, and primordial germ cells during embryogenesis, the proliferation of testicular cells in the postnatal period, and the functions of mature Sertoli and Leydig cells. In addition, p38 MAPK signalling is involved in decreased male fertility when exposed to various harmful stimuli. This review will describe in detail the biological functions of p38 MAPK signalling in testis development and male reproduction, together with its pathological role in male infertility.
Collapse
|
17
|
Källsten L, Almamoun R, Pierozan P, Nylander E, Sdougkou K, Martin JW, Karlsson O. Adult Exposure to Di-N-Butyl Phthalate (DBP) Induces Persistent Effects on Testicular Cell Markers and Testosterone Biosynthesis in Mice. Int J Mol Sci 2022; 23:ijms23158718. [PMID: 35955852 PMCID: PMC9369267 DOI: 10.3390/ijms23158718] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Studies indicate that phthalates are endocrine disruptors affecting reproductive health. One of the most commonly used phthalates, di-n-butyl phthalate (DBP), has been linked with adverse reproductive health outcomes in men, but the mechanisms behind these effects are still poorly understood. Here, adult male mice were orally exposed to DBP (10 or 100 mg/kg/day) for five weeks, and the testis and adrenal glands were collected one week after the last dose, to examine more persistent effects. Quantification of testosterone, androstenedione, progesterone and corticosterone concentrations by liquid chromatography-mass spectrometry showed that testicular testosterone was significantly decreased in both DBP treatment groups, whereas the other steroids were not significantly altered. Western blot analysis of testis revealed that DBP exposure increased the levels of the steroidogenic enzymes CYP11A1, HSD3β2, and CYP17A1, the oxidative stress marker nitrotyrosine, and the luteinizing hormone receptor (LHR). The analysis further demonstrated increased levels of the germ cell marker DAZL, the Sertoli cell markers vimentin and SOX9, and the Leydig cell marker SULT1E1. Overall, the present work provides more mechanistic understanding of how adult DBP exposure can induce effects on the male reproductive system by affecting several key cells and proteins important for testosterone biosynthesis and spermatogenesis, and for the first time shows that these effects persist at least one week after the last dose. It also demonstrates impairment of testosterone biosynthesis at a lower dose than previously reported.
Collapse
|
18
|
Chen H, Chen Q, Zhu Y, Yuan K, Li H, Zhang B, Jia Z, Zhou H, Fan M, Qiu Y, Zhuang Q, Lei Z, Li M, Huang W, Liang L, Yan Q, Wang C. MAP3K1 Variant Causes Hyperactivation of Wnt4/β-Catenin/FOXL2 Signaling Contributing to 46,XY Disorders/Differences of Sex Development. Front Genet 2022; 13:736988. [PMID: 35309143 PMCID: PMC8927045 DOI: 10.3389/fgene.2022.736988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 02/15/2022] [Indexed: 12/28/2022] Open
Abstract
Background: 46,XY disorders/differences of sex development (46,XY DSD) are congenital conditions that result from abnormal gonadal development (gonadal dysgenesis) or abnormalities in androgen synthesis or action. During early embryonic development, several genes are involved in regulating the initiation and maintenance of testicular or ovarian-specific pathways. Recent reports have shown that MAP3K1 genes mediate the development of the 46,XY DSD, which present as complete or partial gonadal dysgenesis. Previous functional studies have demonstrated that some MAP3K1 variants result in the gain of protein function. However, data on possible mechanisms of MAP3K1 genes in modulating protein functions remain scant. Methods: This study identified a Han Chinese family with the 46,XY DSD. To assess the history and clinical manifestations for the 46,XY DSD patients, the physical, operational, ultra-sonographical, pathological, and other examinations were performed for family members. Variant analysis was conducted using both trio whole-exome sequencing (trio WES) and Sanger sequencing. On the other hand, we generated transiently transfected testicular teratoma cells (NT2/D1) and ovary-derived granular cells (KGN), with mutant or wild-type MAP3K1 gene. We then performed functional assays such as determination of steady-state levels of gender related factors, protein interaction and luciferase assay system. Results: Two affected siblings were diagnosed with 46,XY DSD. Our analysis showed a missense c.556A > G/p.R186G variant in the MAP3K1 gene. Functional assays demonstrated that the MAP3K1R186G variant was associated with significantly decreased affinity to ubiquitin (Ub; 43–49%) and increased affinity to RhoA, which was 3.19 ± 0.18 fold, compared to MAP3K1. The MAP3K1R186G led to hyperphosphorylation of p38 and GSK3β, and promoted hyperactivation of the Wnt4/β-catenin signaling. In addition, there was increased recruitment of β-catenin into the nucleus, which enhanced the expression of pro-ovarian transcription factor FOXL2 gene, thus contributing to the 46,XY DSD. Conclusion: Our study identified a missense MAP3K1 variant associated with 46,XY DSD. We demonstrated that MAP3K1R186G variant enhances binding to the RhoA and improves its own stability, resulting in the activation of the Wnt4/β-catenin/FOXL2 pathway. Taken together, these findings provide novel insights into the molecular mechanisms of 46,XY DSD and promotes better clinical evaluation.
Collapse
Affiliation(s)
- Hong Chen
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, China
| | - Qingqing Chen
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yilin Zhu
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Yuan
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huizhu Li
- Department of Pediatrics, Lishui City People’s Hospital, Lishui, China
| | - Bingtao Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zexiao Jia
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Mingjie Fan
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yue Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Qianqian Zhuang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zhaoying Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Mengyao Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, The Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Li Liang
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Chunlin Wang, , Qingfeng Yan, , Li Liang,
| | - Qingfeng Yan
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, China
- *Correspondence: Chunlin Wang, , Qingfeng Yan, , Li Liang,
| | - Chunlin Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Chunlin Wang, , Qingfeng Yan, , Li Liang,
| |
Collapse
|
19
|
Advancements in mammalian X and Y sperm differences and sex control technology. ZYGOTE 2022; 30:423-430. [DOI: 10.1017/s0967199421000939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Summary
Mammal sex determination depends on whether the X sperm or Y sperm binds to the oocyte during fertilization. If the X sperm joins in oocyte, the offspring will be female, if the Y sperm fertilizes, the offspring will be male. Livestock sex control technology has tremendous value for livestock breeding as it can increase the proportion of female offspring and improve the efficiency of livestock production. This review discusses the detailed differences between mammalian X and Y sperm with respect to their morphology, size, and motility in the reproductive tract and in in vitro conditions, as well as ’omics analysis results. Moreover, research progress in mammalian sex control technology has been summarized.
Collapse
|
20
|
Oh M, Son C, Rho SB, Kim M, Park K, Song SY. Stem Cell Factor SOX9 Interacts with a Cell Death Regulator RIPK1 and Results in Escape of Cancer Stem Cell Death. Cells 2022; 11:cells11030363. [PMID: 35159173 PMCID: PMC8834197 DOI: 10.3390/cells11030363] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
High-grade ovarian cancer (HGOC) is the most lethal gynecological cancer, with high metastasis and recurrence. Cancer stem cells (CSCs) are responsible for its apoptosis resistance, cancer metastasis, and recurrence. Thus, targeting CSCs would be a promising strategy for overcoming chemotherapy resistance and improving patient prognosis in HGOC. Among upregulated oncogenic proteins in HGOC, we found that transcription factor SOX9 showed a strong correlation with stemness-regulating ALDH1A1 and was localized predominantly in the cytoplasm of HGOC with lymph node metastasis. In order to address the role of unusual cytoplasmic SOX9 and to explore its underlying mechanism in HGOC malignancy, a Y2H assay was used to identify a necroptotic cell death-associated cytoplasmic protein, receptor-interacting serine/threonine protein kinase 1 (RIPK1), as a novel SOX9-interacting partner and further mapped their respective interacting domains. The C-terminal region containing the transactivation domain of SOX9 interacted with the death domain of R1PK1. Consistent with its stemness-promoting function, SOX9 knockdown in vitro resulted in changes in cell morphology, cell cycle, stem cell marker expression, cell invasion, and sphere formation. Furthermore, in vivo knockdown completely inhibited tumor growth in mouse xenograft model. We propose that cytoplasmic SOX9-mediated cell death suppression would contribute to cancer stem cell survival in HGOC.
Collapse
Affiliation(s)
- Mijung Oh
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Chaeyeon Son
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
| | - Seung Bae Rho
- Division of Translational Science, Research Institute, National Cancer Center, Goyang 10408, Korea;
| | - Minjeong Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Kyoungsook Park
- Medical Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea; (M.O.); (C.S.)
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
- Correspondence: (K.P.); (S.Y.S.); Tel.: +82-10-8718-3625 (K.P.); +82-10-9933-2803 (S.Y.S.)
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
- Correspondence: (K.P.); (S.Y.S.); Tel.: +82-10-8718-3625 (K.P.); +82-10-9933-2803 (S.Y.S.)
| |
Collapse
|
21
|
Granada ML, Audí L. El laboratorio en el diagnóstico multidisciplinar del desarrollo sexual anómalo o diferente (DSD). ADVANCES IN LABORATORY MEDICINE 2021; 2:481-493. [PMCID: PMC10197318 DOI: 10.1515/almed-2020-0119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/24/2021] [Indexed: 06/28/2023]
Abstract
Objetivos El desarrollo de las características sexuales femeninas o masculinas acontece durante la vida fetal, determinándose el sexo genético, el gonadal y el sexo genital interno y externo (femenino o masculino). Cualquier discordancia en las etapas de diferenciación ocasiona un desarrollo sexual anómalo o diferente (DSD) que se clasifica según la composición de los cromosomas sexuales del cariotipo. Contenido En este capítulo se abordan la fisiología de la determinación y el desarrollo de las características sexuales femeninas o masculinas durante la vida fetal, la clasificación general de los DSD y su estudio diagnóstico clínico, bioquímico y genético que debe ser multidisciplinar. Los estudios bioquímicos deben incluir, además de las determinaciones bioquímicas generales, análisis de hormonas esteroideas y peptídicas, en condiciones basales o en pruebas funcionales de estimulación. El estudio genético debe comenzar con la determinación del cariotipo al que seguirá un estudio molecular en los cariotipos 46,XX ó 46,XY, orientado a la caracterización de un gen candidato. Además, se expondrán de manera específica los marcadores bioquímicos y genéticos en los DSD 46,XX, que incluyen el desarrollo gonadal anómalo (disgenesias, ovotestes y testes), el exceso de andrógenos de origen fetal (el más frecuente), fetoplacentario o materno y las anomalías del desarrollo de los genitales internos. Perspectivas El diagnóstico de un DSD requiere la contribución de un equipo multidisciplinar coordinado por un clínico y que incluya los servicios de bioquímica y genética clínica y molecular, un servicio de radiología e imagen y un servicio de anatomía patológica.
Collapse
Affiliation(s)
- Maria Luisa Granada
- Department of Clinical Biochemistry, Hospital Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, España
| | - Laura Audí
- Growth and Development Research Group, Vall d’Hebron Research Institute (VHIR), Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Catalonia, España
| |
Collapse
|
22
|
Granada ML, Audí L. The laboratory in the multidisciplinary diagnosis of differences or disorders of sex development (DSD): I) Physiology, classification, approach, and methodologyII) Biochemical and genetic markers in 46,XX DSD. ADVANCES IN LABORATORY MEDICINE 2021; 2:468-493. [PMID: 37360895 PMCID: PMC10197333 DOI: 10.1515/almed-2021-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/24/2021] [Indexed: 06/28/2023]
Abstract
Objectives The development of female or male sex characteristics occurs during fetal life, when the genetic, gonadal, and internal and external genital sex is determined (female or male). Any discordance among sex determination and differentiation stages results in differences/disorders of sex development (DSD), which are classified based on the sex chromosomes found on the karyotype. Content This chapter addresses the physiological mechanisms that determine the development of female or male sex characteristics during fetal life, provides a general classification of DSD, and offers guidance for clinical, biochemical, and genetic diagnosis, which must be established by a multidisciplinary team. Biochemical studies should include general biochemistry, steroid and peptide hormone testing either at baseline or by stimulation testing. The genetic study should start with the determination of the karyotype, followed by a molecular study of the 46,XX or 46,XY karyotypes for the identification of candidate genes. Summary 46,XX DSD include an abnormal gonadal development (dysgenesis, ovotestes, or testes), an androgen excess (the most frequent) of fetal, fetoplacental, or maternal origin and an abnormal development of the internal genitalia. Biochemical and genetic markers are specific for each group. Outlook Diagnosis of DSD requires the involvement of a multidisciplinary team coordinated by a clinician, including a service of biochemistry, clinical, and molecular genetic testing, radiology and imaging, and a service of pathological anatomy.
Collapse
Affiliation(s)
- Maria Luisa Granada
- Department of Clinical Biochemistry, Hospital Germans Trias i Pujol, Autonomous University of Barcelona, Badalona, Spain
| | - Laura Audí
- Growth and Development Research Group, Vall d’Hebron Research Institute (VHIR), Center for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| |
Collapse
|
23
|
Ridnik M, Schoenfelder S, Gonen N. Cis-Regulatory Control of Mammalian Sex Determination. Sex Dev 2021; 15:317-334. [PMID: 34710870 PMCID: PMC8743899 DOI: 10.1159/000519244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
Sex determination is the process by which an initial bipotential gonad adopts either a testicular or ovarian cell fate. The inability to properly complete this process leads to a group of developmental disorders classified as disorders of sex development (DSD). To date, dozens of genes were shown to play roles in mammalian sex determination, and mutations in these genes can cause DSD in humans or gonadal sex reversal/dysfunction in mice. However, exome sequencing currently provides genetic diagnosis for only less than half of DSD patients. This points towards a major role for the non-coding genome during sex determination. In this review, we highlight recent advances in our understanding of non-coding, cis-acting gene regulatory elements and discuss how they may control transcriptional programmes that underpin sex determination in the context of the 3-dimensional folding of chromatin. As a paradigm, we focus on the Sox9 gene, a prominent pro-male factor and one of the most extensively studied genes in gonadal cell fate determination.
Collapse
Affiliation(s)
- Meshi Ridnik
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Stefan Schoenfelder
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
24
|
Editing SOX Genes by CRISPR-Cas: Current Insights and Future Perspectives. Int J Mol Sci 2021; 22:ijms222111321. [PMID: 34768751 PMCID: PMC8583549 DOI: 10.3390/ijms222111321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 01/16/2023] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and its associated proteins (Cas) is an adaptive immune system in archaea and most bacteria. By repurposing these systems for use in eukaryote cells, a substantial revolution has arisen in the genome engineering field. In recent years, CRISPR-Cas technology was rapidly developed and different types of DNA or RNA sequence editors, gene activator or repressor, and epigenome modulators established. The versatility and feasibility of CRISPR-Cas technology has introduced this system as the most suitable tool for discovering and studying the mechanism of specific genes and also for generating appropriate cell and animal models. SOX genes play crucial roles in development processes and stemness. To elucidate the exact roles of SOX factors and their partners in tissue hemostasis and cell regeneration, generating appropriate in vitro and in vivo models is crucial. In line with these premises, CRISPR-Cas technology is a promising tool for studying different family members of SOX transcription factors. In this review, we aim to highlight the importance of CRISPR-Cas and summarize the applications of this novel, promising technology in studying and decoding the function of different members of the SOX gene family.
Collapse
|
25
|
Jorgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr 2021; 96:190-206. [PMID: 34607330 DOI: 10.1159/000519964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/11/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Male reproductive development in mammals can be divided into a gonadal formation phase followed by a hormone-driven differentiation phase. Failure of these processes may result in Differences in Sex Development (DSD), which may include abnormalities of the male reproductive tract, including cryptorchidism, hypospadias, infertility, and testicular germ cell cancer (TGCC). These disorders are also considered to be part of a testicular dysgenesis syndrome (TDS) in males. Whilst DSDs are considered to result primarily from genetic abnormalities, the development of TDS disorders is frequently associated with environmental factors. SUMMARY In this review, we will discuss the development of the male reproductive system in relation to DSD and TDS. We will also describe the experimental systems, including studies involving animals and human tissues or cells that can be used to investigate the role of environmental factors in inducing male reproductive disorders. We will discuss recent studies investigating the impact of environmental chemicals (e.g., phthalates and bisphenols), lifestyle factors (e.g., smoking) and pharmaceuticals (e.g., analgesics) on foetal testis development. Finally, we will describe the evidence, involving experimental and epidemiologic approaches, for a role of environmental factors in the development of specific male reproductive disorders, including cryptorchidism, hypospadias, and TGCC. KEY MESSAGES Environmental exposures can impact the development and function of the male reproductive system in humans. Epidemiology studies and experimental approaches using human tissues are important to translate findings from animal studies and account for species differences in response to environmental exposures.
Collapse
Affiliation(s)
- Anne Jorgensen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Harriet Miles
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Tarini Chetty
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Rod T Mitchell
- Royal Hospital for Children and Young People, Edinburgh, UK
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
26
|
Hickmann FMW, Braccini Neto J, Kramer LM, Huang Y, Gray KA, Dekkers JCM, Sanglard LP, Serão NVL. Host Genetics of Response to Porcine Reproductive and Respiratory Syndrome in Sows: Reproductive Performance. Front Genet 2021; 12:707870. [PMID: 34422010 PMCID: PMC8371709 DOI: 10.3389/fgene.2021.707870] [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] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/07/2021] [Indexed: 11/13/2022] Open
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) is historically the most economically important swine disease worldwide that severely affects the reproductive performance of sows. However, little is still known about the genetic basis of reproductive performance in purebred herds during a PRRS outbreak through the comparison of maternal and terminal breeds. Thus, the objective of this work was to explore the host genetics of response to PRRS in purebred sows from two breeds. Reproductive data included 2546 Duroc and 2522 Landrace litters from 894 and 813 purebred sows, respectively, which had high-density genotype data available (29,799 single nucleotide polymorphisms; SNPs). The data were split into pre-PRRS, PRRS, and post-PRRS phases based on standardized farrow-year-week estimates. Heritability estimates for reproductive traits were low to moderate (≤0.20) for Duroc and Landrace across PRRS phases. On the other hand, genetic correlations of reproductive traits between PRRS phases were overall moderate to high for both breeds. Several associations between MARC0034894, a candidate SNP for response to PRRS, with reproductive performance were identified (P-value < 0.05). Genomic analyses detected few QTL for reproductive performance across all phases, most explaining a small percentage of the additive genetic variance (≤8.2%, averaging 2.1%), indicating that these traits are highly polygenic. None of the identified QTL within a breed and trait overlapped between PRRS phases. Overall, our results indicate that Duroc sows are phenotypically more resilient to PRRS than Landrace sows, with a similar return to PRRS-free performance between breeds for most reproductive traits. Genomic prediction results indicate that genomic selection for improved reproductive performance under a PRRS outbreak is possible, especially in Landrace sows, by training markers using data from PRRS-challenged sows. On the other hand, the high genetic correlations with reproductive traits between PRRS phases suggest that selection for improved reproductive performance in a clean environment could improve performance during PRRS, but with limited efficiency due to their low heritability estimates. Thus, we hypothesize that an indicator trait that could be indirectly selected to increase the response to selection for these traits would be desirable and would also improve the reproductive performance of sows during a PRRS outbreak.
Collapse
Affiliation(s)
- Felipe M. W. Hickmann
- Department of Animal Science, Iowa State University, Ames, IA, United States
- Department of Animal Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - José Braccini Neto
- Department of Animal Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luke M. Kramer
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Yijian Huang
- Smithfield Premium Genetics, Rose Hill, NC, United States
| | - Kent A. Gray
- Smithfield Premium Genetics, Rose Hill, NC, United States
| | - Jack C. M. Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Leticia P. Sanglard
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Nick V. L. Serão
- Department of Animal Science, Iowa State University, Ames, IA, United States
| |
Collapse
|
27
|
Han F, Yin L, Jiang X, Zhang X, Zhang N, Yang J, Ouyang W, Hao X, Liu W, Huang Y, Chen H, Gao F, Li Z, Guo Q, Cao J, Liu J. Identification of SRY-box 30 as an age-related essential gatekeeper for male germ-cell meiosis and differentiation. Aging Cell 2021; 20:e13343. [PMID: 33721419 PMCID: PMC8135013 DOI: 10.1111/acel.13343] [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] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/07/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022] Open
Abstract
Although important factors governing the meiosis have been reported in the embryonic ovary, meiosis in postnatal testis remains poorly understood. Herein, we first report that SRY‐box 30 (Sox30) is an age‐related and essential regulator of meiosis in the postnatal testis. Sox30‐null mice exhibited uniquely impaired testis, presenting the abnormal arrest of germ‐cell differentiation and irregular Leydig cell proliferation. In aged Sox30‐null mice, the observed testicular impairments were more severe. Furthermore, the germ‐cell arrest occurred at the stage of meiotic zygotene spermatocytes, which is strongly associated with critical regulators of meiosis (such as Cyp26b1, Stra8 and Rec8) and sex differentiation (such as Rspo1, Foxl2, Sox9, Wnt4 and Ctnnb1). Mechanistically, Sox30 can activate Stra8 and Rec8, and inhibit Cyp26b1 and Ctnnb1 by direct binding to their promoters. A different Sox30 domain required for regulating the activity of these gene promoters, providing a “fail‐safe” mechanism for Sox30 to facilitate germ‐cell differentiation. Indeed, retinoic acid levels were reduced owing to increased degradation following the elevation of Cyp26b1 in Sox30‐null testes. Re‐expression of Sox30 in Sox30‐null mice successfully restored germ‐cell meiosis, differentiation and Leydig cell proliferation. Moreover, the restoration of actual fertility appeared to improve over time. Consistently, Rec8 and Stra8 were reactivated, and Cyp26b1 and Ctnnb1 were reinhibited in the restored testes. In summary, Sox30 is necessary, sufficient and age‐associated for germ‐cell meiosis and differentiation in testes by direct regulating critical regulators. This study advances our understanding of the regulation of germ‐cell meiosis and differentiation in the postnatal testis.
Collapse
Affiliation(s)
- Fei Han
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Li Yin
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
- College of Pharmacy and Bioengineering Chongqing University of Technology Chongqing China
| | - Xiao Jiang
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Xi Zhang
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Ning Zhang
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Jun‐tang Yang
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
- College of Life Science Henan Normal University Henan China
| | - Wei‐ming Ouyang
- Office of Biotechnology Products Center for Drug Evaluation and Research U.S. Food and Drug Administration Pittsburgh PA USA
| | - Xiang‐lin Hao
- Department of Pathology Xinqiao HospitalArmy Medical University Chongqing China
| | - Wen‐bin Liu
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Yong‐sheng Huang
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Hong‐qiang Chen
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Fei Gao
- Department of Veterinary and Animal Sciences Faculty of Health and Medical Sciences University of Copenhagen Frederiksberg DK Denmark
| | - Zhong‐tai Li
- Department of Urology Daping HospitalArmy Medical University Chongqing China
| | - Qiao‐nan Guo
- Department of Pathology Xinqiao HospitalArmy Medical University Chongqing China
| | - Jia Cao
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| | - Jin‐yi Liu
- Institute of Toxicology College of Preventive Medicine Army Medical University Chongqing China
| |
Collapse
|
28
|
Frost ER, Taylor G, Baker MA, Lovell-Badge R, Sutherland JM. Establishing and maintaining fertility: the importance of cell cycle arrest. Genes Dev 2021; 35:619-634. [PMID: 33888561 PMCID: PMC8091977 DOI: 10.1101/gad.348151.120] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this review, Frost et al. summarize the current knowledge on the Cip/Kip family of cyclin-dependent kinase inhibitors in mouse gonad development and highlight new roles for cell cycle inhibitors in controlling and maintaining female fertility. Development of the ovary or testis is required to establish reproductive competence. Gonad development relies on key cell fate decisions that occur early in embryonic development and are actively maintained. During gonad development, both germ cells and somatic cells proliferate extensively, a process facilitated by cell cycle regulation. This review focuses on the Cip/Kip family of cyclin-dependent kinase inhibitors (CKIs) in mouse gonad development. We particularly highlight recent single-cell RNA sequencing studies that show the heterogeneity of cyclin-dependent kinase inhibitors. This diversity highlights new roles for cell cycle inhibitors in controlling and maintaining female fertility.
Collapse
Affiliation(s)
- Emily R Frost
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia.,Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Güneş Taylor
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| |
Collapse
|
29
|
Vining B, Ming Z, Bagheri-Fam S, Harley V. Diverse Regulation but Conserved Function: SOX9 in Vertebrate Sex Determination. Genes (Basel) 2021; 12:genes12040486. [PMID: 33810596 PMCID: PMC8066042 DOI: 10.3390/genes12040486] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/15/2022] Open
Abstract
Sex determination occurs early during embryogenesis among vertebrates. It involves the differentiation of the bipotential gonad to ovaries or testes by a fascinating diversity of molecular switches. In most mammals, the switch is SRY (sex determining region Y); in other vertebrates it could be one of a variety of genes including Dmrt1 or dmy. Downstream of the switch gene, SOX9 upregulation is a central event in testes development, controlled by gonad-specific enhancers across the 2 Mb SOX9 locus. SOX9 is a ‘hub’ gene of gonadal development, regulated positively in males and negatively in females. Despite this diversity, SOX9 protein sequence and function among vertebrates remains highly conserved. This article explores the cellular, morphological, and genetic mechanisms initiated by SOX9 for male gonad differentiation.
Collapse
Affiliation(s)
- Brittany Vining
- Sex Development Laboratory, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (B.V.); (Z.M.); (S.B.-F.)
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC 3800, Australia
| | - Zhenhua Ming
- Sex Development Laboratory, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (B.V.); (Z.M.); (S.B.-F.)
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC 3800, Australia
| | - Stefan Bagheri-Fam
- Sex Development Laboratory, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (B.V.); (Z.M.); (S.B.-F.)
| | - Vincent Harley
- Sex Development Laboratory, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; (B.V.); (Z.M.); (S.B.-F.)
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC 3800, Australia
- Correspondence: ; Tel.: +61-3-8572-2527
| |
Collapse
|
30
|
Park JH, Kim YH, Shim S, Kim A, Jang H, Lee SJ, Park S, Seo S, Jang WI, Lee SB, Kim MJ. Radiation-Activated PI3K/AKT Pathway Promotes the Induction of Cancer Stem-Like Cells via the Upregulation of SOX2 in Colorectal Cancer. Cells 2021; 10:cells10010135. [PMID: 33445526 PMCID: PMC7827893 DOI: 10.3390/cells10010135] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
The current treatment strategy for patients with aggressive colorectal cancer has been hampered by resistance to radiotherapy and chemotherapy due to the existence of cancer stem-like cells (CSCs). Recent studies have shown that SOX2 expression plays an important role in the maintenance of CSC properties in colorectal cancer. In this study, we investigated the induction and regulatory role of SOX2 following the irradiation of radioresistant and radiosensitive colorectal cancer cells. We used FACS and western blotting to analyze SOX2 expression in cells. Among the markers of colorectal CSCs, the expression of CD44 increased upon irradiation in radioresistant cells. Further analysis revealed the retention of CSC properties with an upregulation of SOX2 as shown by enhanced resistance to radiation and metastatic potential in vitro. Interestingly, both the knockdown and overexpression of SOX2 led to increase in CD44+ population and induction of CSC properties in colorectal cancer following irradiation. Furthermore, selective genetic and pharmacological inhibition of the PI3K/AKT pathway, but not the MAPK pathway, attenuated SOX2-dependent CD44 expression and metastatic potential upon irradiation in vitro. Our findings suggested that SOX2 regulated by radiation-induced activation of PI3K/AKT pathway contributes to the induction of colorectal CSCs, thereby highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Ji-Hye Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Young-Heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Areumnuri Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 04763, Korea;
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Songwon Seo
- Laboratory of Radiation Epidermiology, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Won Il Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
- Correspondence: (S.B.L.); (M.-J.K.); Tel.: +82-2-3399-5874 (S.B.L.); +82-2-3399-5875 (M.-J.K.)
| | - Min-Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (J.-H.P.); (Y.-H.K.); (S.S.); (A.K.); (H.J.); (S.P.); (W.I.J.)
- Correspondence: (S.B.L.); (M.-J.K.); Tel.: +82-2-3399-5874 (S.B.L.); +82-2-3399-5875 (M.-J.K.)
| |
Collapse
|
31
|
Moradi M, Hajarian H, Karamishabankareh H, Soltani L, Soleymani B. Recovery of sperms bearing X chromosomes with different concentrations of magnetic nanoparticles in ram. Reprod Domest Anim 2020; 56:263-269. [PMID: 32813917 DOI: 10.1111/rda.13807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/28/2020] [Accepted: 08/15/2020] [Indexed: 11/29/2022]
Abstract
Pre-conceptual sex selection is still a highly debatable process whereby X and Y chromosome bearing spermatozoa are isolated before oocyte fertilization. Recently, magnetic nanoparticles (MNP) have been used to determine X and Y chromosomes bearing spermatozoa as a result of searching for a cheap, highly efficient method using non-toxic materials. This study aimed to recover the sperm bearing X chromosomes in ram with different concentrations of MNP and then evaluate the success of this method using polymerase chain reaction (PCR). Ram sperms were divided into four groups, treated with 0 (control), 50, 100 and 200 μg/ml MNP, respectively. MNP was used to restore sperm cells bearing X chromosomes. Upon recovery, the PCR was performed to identify the X and Y sperms, Methyl ThiazoleTetrazolium (MTT), to assess MNP toxicity and sperm viability and acridine orange (AO) to evaluate sperm DNA integrity. The results of PCR revealed that the treatment of spermatozoa- bearing X chromosomes with 50 μg/ml MNP had the highest effects on the recovery of X sperm rather than the other concentrations of MNP. However, the concentrations of MNP did not have any toxic effects on spermatozoa, sperm viability and, DNA integrity, but the high concentration of MNP (200 μg/ml) significantly reduced DNA integrity. According to MTT and AO results, the concentrations of MNP used in this study had no toxic effects on spermatozoa and did not reduce the sperm viability and DNA integrity, except that 200 μg/ml MNP significantly reduced DNA integrity.
Collapse
Affiliation(s)
- Maryam Moradi
- Department of Animal Science, Faculty of Agricultural and Engineering Science, Razi University, Kermanshah, Iran
| | - Hadi Hajarian
- Department of Animal Science, Faculty of Agricultural and Engineering Science, Razi University, Kermanshah, Iran
| | - Hamed Karamishabankareh
- Department of Animal Science, Faculty of Agricultural and Engineering Science, Razi University, Kermanshah, Iran
| | - Leila Soltani
- Department of Animal Science, Faculty of Agricultural and Engineering Science, Razi University, Kermanshah, Iran
| | - Bijan Soleymani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Science, Kermanshah, Iran
| |
Collapse
|
32
|
Alikhani M, Karamzadeh R, Rahimi P, Adib S, Baharvand H, Salekdeh GH. Human Proteome Project and Human Pluripotent Stem Cells: Odd Bedfellows or a Perfect Match? J Proteome Res 2020; 19:4747-4753. [PMID: 33124832 DOI: 10.1021/acs.jproteome.0c00689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The Chromosome-Centric Human Proteome Project (C-HPP) aims at the identification of missing proteins (MPs) and the functional characterization of functionally unannotated PE1 (uPE1) proteins. A major challenge in addressing this goal is that many human proteins and MPs are silent in adult cells. A promising approach to overcome such challenge is to exploit the advantage of novel tools such as pluripotent stem cells (PSCs), which are capable of differentiation into three embryonic germ layers, namely, the endoderm, mesoderm, and ectoderm. Here we present several examples of how the Human Y Chromosome Proteome Project (Y-HPP) benefited from this approach to meet C-HPP goals. Furthermore, we discuss how integrating CRISPR engineering, human-induced pluripotent stem cell (hiPSC)-derived disease modeling systems, and organoid technologies provides a unique platform for Y-HPP and C-HPP for MP identification and the functional characterization of human proteins, especially uPE1s.
Collapse
Affiliation(s)
- Mehdi Alikhani
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Razieh Karamzadeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Pardis Rahimi
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Samane Adib
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115-111, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 146196815, Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Molecular Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
33
|
Stewart MK, Mattiske DM, Pask AJ. Oestrogen regulates SOX9 bioavailability by rapidly activating ERK1/2 and stabilising microtubules in a human testis-derived cell line. Exp Cell Res 2020; 398:112405. [PMID: 33271127 DOI: 10.1016/j.yexcr.2020.112405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 01/31/2023]
Abstract
Nuclear SOX9 is essential for Sertoli cell differentiation and the development of a testis. Exposure of Sertoli cells to exogenous oestrogen causes cytoplasmic retention of SOX9, inhibiting testis development and promoting ovarian development. The cytoplasmic localisation of SOX9 requires a stabilised microtubule network and a key MAPK complex, ERK1/2, is responsive to oestrogen and known to affect the microtubule network. We hypothesised that oestrogen could stabilise microtubules through the activation of ERK1/2 to promote the cytoplasmic retention of SOX9. Treatment of human testis-derived NT2/D1 cells for 30 min with oestrogen rapidly activated ERK1/2, stabilised the microtubule network and increased cytoplasmic localisation of SOX9. The effects of oestrogen on SOX9 and tubulin were blocked by the ERK1/2 inhibitor U0126, demonstrating that ERK1/2 mediates the stabilisation of microtubules and cytoplasmic retention of SOX9 by oestrogen. Together, these data revealed a previously unknown mechanism for oestrogen in impacting MAPK signalling to block SOX9 bioavailability and the differentiation of Sertoli cells.
Collapse
Affiliation(s)
- Melanie K Stewart
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Deidre M Mattiske
- School of BioSciences, The University of Melbourne, Victoria, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Victoria, Australia.
| |
Collapse
|
34
|
de la Calle CM, Kim S, Baskin LS. Diagnosis and treatment of the intra-abdominal gonad in the pediatric population: Testes, ovaries, dysgenetic gonads, streaks, and ovotestes. J Pediatr Surg 2020; 55:2480-2491. [PMID: 32164982 DOI: 10.1016/j.jpedsurg.2020.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/14/2020] [Accepted: 02/16/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND/PURPOSE Pediatric surgical specialists are often confronted with the difficult task of identifying, diagnosing and managing intra-abdominal gonads in children. Ranging from the intra-abdominal cryptorchid testis to normal or pathologic ovaries and gonads in disorders of sexual development, all intra-abdominal gonads in the pediatric population pose different diagnosis and management challenges. Understanding the hormonal and fertility potential of the gonad and knowing its potential cancer risk is essential when deciding how to manage these patients. In addition, the ideal surgical management for each one of these patients is often debated. METHODS Descriptive literature review. RESULTS/CONCLUSION Herein, we reviewed gonadal formation, common etiologies, diagnosis and management of intra-abdominal testes, pathologic ovaries and gonads in disorders of sexual development. Fertility potential and cancer risk for each were also reviewed and how both affect surgical management of the gonad. TYPE OF STUDY/LEVEL OF EVIDENCE Review Article, Level V.
Collapse
Affiliation(s)
- Claire M de la Calle
- Department of Urology, University of California San Francisco, San Francisco, CA, USA.
| | - Sunghoon Kim
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Laurence S Baskin
- Department of Urology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
35
|
Vallarino N, Wydooghe E, van Goethem B. Laparoscopic gonadectomy in dogs with ovotesticular disorder of sexual development. Reprod Domest Anim 2020; 55:1172-1179. [PMID: 32599672 DOI: 10.1111/rda.13759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/20/2020] [Indexed: 11/30/2022]
Abstract
Disorders of sexual development (DSD) in dogs involve most commonly an XX sex reversal syndrome, treated conventionally by gonadohysterectomy. The objective of the present case series is to describe the surgical treatment and long-term follow-up of dogs undergoing laparoscopic gonadectomy without hysterectomy for treatment of ovotesticular DSD. Six female dogs clinically diagnosed with DSD were retrospectively included in the study when laparoscopic gonadectomy was performed and histology confirmed the presence of abnormal gonads. The dogs were evaluated by ultrasound after 6 months, and owners were contacted by phone for the long-term reevaluation. Laparoscopic gonadectomy was performed using 2- or 3-portal midline techniques with 3- and/or 5-mm instruments. Additional procedures were performed in 5 dogs, including os clitoris removal in 4 dogs and vulvoplasty in 1 dog. Histological analysis of the gonads reported 11 ovotestes and 1 testis. No major or minor complications occurred perioperatively. Ultrasonographic reevaluation was performed in 5/6 dogs and the remaining abdominal genital system was considered normal. Median long-term follow-up was 617 days (range, 265-1597) with none of the dogs having any symptom related to DSD. Therefore, laparoscopic gonadectomy is a valid alternative for dogs with ovotesticular DSD and is less invasive than conventional open techniques. Removal of the gonads avoids future development of hormone-related diseases of the remaining genital tract.
Collapse
Affiliation(s)
- Nicolas Vallarino
- Small Animal Teaching Hospital, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eline Wydooghe
- Department of Obstetrics, Reproduction and Herd Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart van Goethem
- Small Animal Teaching Hospital, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
36
|
Awogbindin IO, Adedara IA, Adeniyi PA, Agedah AE, Oyetunde BF, Olorunkalu PD, Ogbuewu E, Akindoyeni IA, Mustapha YE, Ezekiel OG, Farombi EO. Nigral and ventral tegmental area lesioning induces testicular and sperm morphological abnormalities in a rotenone model of Parkinson's disease. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103412. [PMID: 32439558 DOI: 10.1016/j.etap.2020.103412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/26/2020] [Accepted: 05/12/2020] [Indexed: 06/11/2023]
Abstract
Although sexual health is affected by Parkinson's disease (PD), the effect on testicular health and/or sperm quality is not well discussed. After 21 days of rotenone lesioning, we observed dopaminergic neuronal degeneration in the substantia nigra and hypothalamus. There were minimal SPACA-1-expressing epididymal spermatozoa with morphological abnormalities, scanty luminal spermatozoa and reduced testicular spermatids and post-meiotic germ cells indicating hypospermatogenesis. Occludin-expressing sertoli cells were dispersed over a wide area indicating compromised blood-testes barrier. Activated caspase-3 expression was intense while immunoreactivity of spermatogenic-enhancing SRY and GADD45 g was weak. Although serum follicle stimulating hormone level was not affected, the lesion was associated with reduced serum testosterone level, testicular oxidative damage and inhibition of acetylcholinesterase activity, even when rotenone was not detected in the testes. Together, dopaminergic lesions may mediate testicular and sperm abnormalities via the brain-hypothalamic-testicular circuit independent of the pituitary, thereby establishing a causal link between Parkinsonism and reproductive dysfunction.
Collapse
Affiliation(s)
- Ifeoluwa O Awogbindin
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Isaac A Adedara
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Philip A Adeniyi
- Cell Biology and Neurotoxicity Unit, Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
| | - Alberta E Agedah
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Bisola F Oyetunde
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Precious D Olorunkalu
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Emmanuel Ogbuewu
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Inioluwa A Akindoyeni
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Yusuf E Mustapha
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oluwatoyin G Ezekiel
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ebenezer O Farombi
- Drug Metabolism and Molecular Toxicology Research Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
37
|
Quan MY, Guo Q, Liu J, Yang R, Bai J, Wang W, Cai Y, Han R, Lv YQ, Ding L, Billadeau DD, Lou Z, Bellusci S, Li X, Zhang JS. An FGFR/AKT/SOX2 Signaling Axis Controls Pancreatic Cancer Stemness. Front Cell Dev Biol 2020; 8:287. [PMID: 32457900 PMCID: PMC7221133 DOI: 10.3389/fcell.2020.00287] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/02/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer stemness is associated with high malignancy and low differentiation, as well as therapeutic resistance of tumors including pancreatic ductal adenocarcinoma (PDAC). Fibroblast growth factors (FGFs) exert pleiotropic effects on a variety of cellular processes and functions including embryonic stem cell pluripotency and cancer cell stemness via the activation of four tyrosine kinase FGF receptors (FGFRs). FGF ligands have been a major component of the cocktail of growth factors contained in the cancer stemness-inducing (CSI) and organoid culture medium. Although FGF/FGFR signaling has been hypothesized to maintain cancer stemness, its function in this process is still unclear. We report that inhibition of FGF/FGFR signaling impairs sphere-forming ability of PDAC in vitro, and knocking down FGFR1 and FGFR2 decreased their tumorigenesis abilities in vivo. Mechanistically, we demonstrated that SOX2 is down-regulated upon loss of FGFR signaling. The overexpression of SOX2 in SOX2-negative cells, which normally do not display stemness capabilities, is sufficient to induce spheroid formation. Additionally, we found that AKT phosphorylation was reduced upon FGFR signaling inhibition. The inhibition of AKT using specific pharmacological inhibitors in the context of CSI medium leads to the loss of spheroid formation associated with loss of SOX2 nuclear expression and increased degradation. We demonstrate that an FGFR/AKT/SOX2 axis controls cancer stemness in PDAC and therefore may represent an important therapeutic target in the fight against this very aggressive form of cancer.
Collapse
Affiliation(s)
- Mei-Yu Quan
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Qiang Guo
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jiayu Liu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Ruo Yang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jing Bai
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Yaxin Cai
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Rui Han
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Yu-Qing Lv
- Center for Precision Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Ding
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Daniel D Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Zhenkun Lou
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Saverio Bellusci
- Institute of Life Sciences, Wenzhou University, Wenzhou, China.,Cardio-Pulmonary Institute, Member of the German Lung Center, Justus Liebig University Giessen, Giessen, Germany
| | - Xiaokun Li
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jin-San Zhang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China.,Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
38
|
aarF domain containing kinase 5 gene promotes invasion and migration of lung cancer cells through ADCK5-SOX9-PTTG1 pathway. Exp Cell Res 2020; 392:112002. [PMID: 32277958 DOI: 10.1016/j.yexcr.2020.112002] [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: 12/23/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/08/2023]
Abstract
AarF domain containing kinase 5 (ADCK5) is a member of an atypical kinase family and overexpressed in many carcinomas including lung cancer, while the function of this protein has not been elucidated. Here we investigated the mechanism of ADCK5 involved in regulating invasion and migration of lung cancer cells, and showed that ADCK5 might regulate the expression of tumor oncogene human pituitary tumor transforming gene-1 (PTTG1) by phosphorylating transcription factor SOX9, therefore enhancing the migration and invasion capabilities of lung cancer cells. Mutagenesis of potential serine phosphorylation sites on SOX9 indicated that serine 181 might be required to maintain transcription activation of SOX9 as well as increase PTTG1 levels. The serine 181 site of SOX9 is in a motif that is targeted by ADCK5. The ADCK5-SOX9-PTTG1 pathway might be a potential therapeutic target for lung cancer.
Collapse
|
39
|
Disorders of Sex Development-Novel Regulators, Impacts on Fertility, and Options for Fertility Preservation. Int J Mol Sci 2020; 21:ijms21072282. [PMID: 32224856 PMCID: PMC7178030 DOI: 10.3390/ijms21072282] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Disorders (or differences) of sex development (DSD) are a heterogeneous group of congenital conditions with variations in chromosomal, gonadal, or anatomical sex. Impaired gonadal development is central to the pathogenesis of the majority of DSDs and therefore a clear understanding of gonadal development is essential to comprehend the impacts of these disorders on the individual, including impacts on future fertility. Gonadal development was traditionally considered to involve a primary 'male' pathway leading to testicular development as a result of expression of a small number of key testis-determining genes. However, it is increasingly recognized that there are several gene networks involved in the development of the bipotential gonad towards either a testicular or ovarian fate. This includes genes that act antagonistically to regulate gonadal development. This review will highlight some of the novel regulators of gonadal development and how the identification of these has enhanced understanding of gonadal development and the pathogenesis of DSD. We will also describe the impact of DSDs on fertility and options for fertility preservation in this context.
Collapse
|
40
|
García-Acero M, Moreno O, Suárez F, Rojas A. Disorders of Sexual Development: Current Status and Progress in the Diagnostic Approach. Curr Urol 2020; 13:169-178. [PMID: 31998049 DOI: 10.1159/000499274] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022] Open
Abstract
Disorders of sexual development (DSD) are conditions with an atypical chromosomal, gonadal or phenotypic sex, which leads to differences in the development of the urogenital tract and different clinical phenotypes. Some genes have been implicated in the sex development during gonadal and functional differentiation where the maintenance of the somatic sex of the gonad as either male or female is achieved by suppression of the alternate route. The diagnosis of DSD requires a structured approach, involving a multidisciplinary team and different molecular techniques. We discuss the dimorphic genes and the specific pathways involved in gonadal differentiation, as well as new techniques for genetic analysis and their diagnostic value including epigenetic mechanisms, expanding the evidence in the diagnostic approach of individuals with DSD to increase knowledge of the etiology.
Collapse
Affiliation(s)
- Mary García-Acero
- Human Genetic Institute, Medicine Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Olga Moreno
- Human Genetic Institute, Medicine Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Fernando Suárez
- Human Genetic Institute, Medicine Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Rojas
- Human Genetic Institute, Medicine Faculty, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
41
|
Taleahmad S, Alikhani M, Mollamohammadi S, Yousefi M, Taei A, Hassani SN, Baharvand H, Salekdeh GH. Inhibition of Human Y Chromosome Gene, SRY, Promotes Naïve State of Human Pluripotent Stem Cells. J Proteome Res 2019; 18:4254-4261. [PMID: 31580082 DOI: 10.1021/acs.jproteome.9b00396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Although males and females have a variety of sexually dimorphic features related to hormonal effects, the genetic basis of dimorphism relies on early embryo development. Two pluripotent states, naïve and primed, emerge during early mammalian development. Identification of signaling pathways that induce differences between these two states can help to modulate conversion of primed cells to naïve cells. Naïve cells have a shorter doubling time and longer survival than their primed counterparts when passaged as single cells. In this study, we sought to explore the role of Y chromosome genes on human pluripotent stem cells (hPSCs) by investigating differential expressions of the male-specific region of the Y chromosome (MSY) genes in primed and naïve cells. Interestingly, we found that several MSY genes, including SRY, showed higher expression levels in primed compared to naïve human embryonic stem cells (hESCs). We hypothesize that SRY prevents WNT/β-catenin signaling by its interaction and inhibition of β-catenin activation in the nucleus. Results of the loss-of-function approach conducted by depletion of SRY indicated increased expressions of pluripotency marker genes and alkaline phosphatase (ALP) activity in the primed cells. SRY reduction was associated with overexpression of WNT signaling target genes AXIN2, Brachury, TCF1, TBX2, and TBX3. We suggest that inhibition of SRY may result in activation of β-catenin and up-regulation of the WNT signaling pathway, both of which are important to naïve conversion.
Collapse
Affiliation(s)
- Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Mehdi Alikhani
- Department of Molecular Systems Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Sepideh Mollamohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Meisam Yousefi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Seyedeh Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
- Department of Developmental Biology , University of Science and Culture , Tehran 113145-871 , Iran
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran 16635-148 , Iran
- Department of Molecular Sciences , Macquarie University , Sydney , NSW 2109 , Australia
- Department of Systems and Synthetic Biology , Agricultural Biotechnology Research Institute of Iran , Karaj 313593315 , Iran
| |
Collapse
|
42
|
Washio K, Mizushima S, Jogahara T, Kuroiwa A. Regulation of the Sox3 Gene in an X0/X0 Mammal without Sry, the Amami Spiny Rat, Tokudaia osimensis. Cytogenet Genome Res 2019; 159:143-150. [PMID: 31760386 DOI: 10.1159/000504313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2019] [Indexed: 11/19/2022] Open
Abstract
Two species of spiny rats, Tokudaia osimensis and Tokudaia tokunoshimensis, show an X0/X0 sex chromosome constitution due to the lack of a Y chromosome. The Sry gene has been completely lost from the genome of these species. We hypothesized that Sox3, which is thought to be originally a homologue of Sry, could function in sex determination in these animals in the absence of Sry. Sox3 was localized in a region of the X chromosome in T. osimensis homologous to mouse. A similar testis- and ovary-specific pattern of expression was observed in mouse and T. osimensis. Although the sequence of the Sox3 gene and its promoter are highly conserved, a 13-bp deletion was specifically found in the promoter region of the 2 spiny rat species. Reporter gene assays were performed to examine the effect of the 13-bp deletion in the promoter region on Sox3 regulation. Although an approximately 60% decrease in activity was observed using the Tokudaia promoters with the 13-bp deletion, the activity was recovered using a mutated promoter in which the deletion was filled with mouse sequence. To evaluate whether SOX3 could regulate Sox9 expression, a reporter gene assay was carried out using testis-specific enhancer of Sox9 core (TESCO). Co-transfection with a combination of mouse SF1 and mouse SOX3 or T. osimensis SOX3 resulted in a greater than 2-fold increase in activity of mouse and T. osimensis TESCO. These results support the idea that the function of SOX3 as a transcription factor, as has been reported in mice and humans, is conserved in T. osimensis. Therefore, we conclude that the Sox3 gene has no function in sex determination in Sry-lacking Tokudaia species.
Collapse
|
43
|
Okashita N, Kuroki S, Maeda R, Tachibana M. TET2 catalyzes active DNA demethylation of the Sry promoter and enhances its expression. Sci Rep 2019; 9:13462. [PMID: 31530896 PMCID: PMC6748950 DOI: 10.1038/s41598-019-50058-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/05/2019] [Indexed: 12/30/2022] Open
Abstract
SRY is the master regulator of male sex determination in eutherian mammals. In mice, Sry expression is transcriptionally and epigenetically controlled in a developmental stage-specific manner. The Sry promoter undergoes demethylation in embryonic gonadal somatic cells at the sex-determining period. However, its molecular mechanism and in vivo significance remain unclear. Here, we report that the Sry promoter is actively demethylated during gonadal development, and TET2 plays a fundamental role in Sry demethylation. Tet2-deficient mice showed absence of 5-hydroxymethylcytosine in the Sry promoter. Furthermore, Tet2 deficiency diminished Sry expression, indicating that TET2-mediated DNA demethylation regulates Sry expression positively. We previously showed that the deficiency of the H3K9 demethylase Jmjd1a compromises Sry expression and induces male-to-female sex reversal. Tet2 deficiency enhanced the sex reversal phenotype of Jmjd1a-deficient mice. Thus, TET2-mediated active DNA demethylation and JMJD1A-mediated H3K9 demethylation contribute synergistically to sex determination.
Collapse
Affiliation(s)
- Naoki Okashita
- Division of Epigenome Dynamics, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan.,Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shunsuke Kuroki
- Division of Epigenome Dynamics, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan.,Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryo Maeda
- Division of Epigenome Dynamics, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan.,Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Tachibana
- Division of Epigenome Dynamics, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan. .,Laboratory of Epigenome Dynamics, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
44
|
Du J, Liu Y, Song C, Cui Z. Discovery of sex-related genes from embryonic development stage based on transcriptome analysis in Eriocheir sinensis. Gene 2019; 710:1-8. [DOI: 10.1016/j.gene.2019.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/27/2019] [Accepted: 05/08/2019] [Indexed: 01/10/2023]
|
45
|
Ogata Y, Nishikata M, Kitada K, Mizushima S, Jogahara T, Kuroiwa A. Spiny rat SRY lacks a long Q-rich domain and is not stable in transgenic mice. Dev Dyn 2019; 248:784-794. [PMID: 31219647 DOI: 10.1002/dvdy.73] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Although Tokudaia muenninki has multiple extra copies of the Sry gene on the Y chromosome, loss of function of these sequences is indicated. To examine the Sry gene function for sex determining in T. muenninki, we screened a BAC library and identified a clone (SRY26) containing complete SRY coding and promoter sequences. RESULTS SRY26 showed high identity to mouse and rat SRY. In an in vitro reporter gene assay, SRY26 was unable to activate testis-specific enhancer of Sox9. Four lines of BAC transgenic mice carrying SRY26 were generated. Although the embryonic gonads of XX transgenic mice displayed sufficient expression levels of SRY26 mRNA, these mice exhibited normal female phenotypes in the external and internal genitalia, and up-regulation of Sox9 was not observed. Expression of the SRY26 protein was confirmed in primate-derived COS7 cells transfected with a SRY26 expression vector. However, the SRY26 protein was not expressed in the gonads of BAC transgenic mice. CONCLUSIONS Overall, these results support a previous study demonstrated a long Q-rich domain plays essential roles in protein stabilization in mice. Therefore, the original aim of this study, to examine the function of the Sry gene of this species, was not achieved by creating TG mice.
Collapse
Affiliation(s)
- Yuka Ogata
- Biosystems Science Course, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mana Nishikata
- Biosystems Science Course, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kazuhiro Kitada
- Biosystems Science Course, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan.,Division of Reproductive and Developmental Biology, Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shusei Mizushima
- Biosystems Science Course, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan.,Division of Reproductive and Developmental Biology, Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takamichi Jogahara
- Division of Bio-Resources, Frontier Science Research Center, Kiyotake Campus, University of Miyazaki, Miyazaki, Japan.,Department of Law and Economics, Okinawa University, Naha, Okinawa, Japan
| | - Asato Kuroiwa
- Biosystems Science Course, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido, Japan.,Division of Reproductive and Developmental Biology, Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
46
|
Aguilar-Medina M, Avendaño-Félix M, Lizárraga-Verdugo E, Bermúdez M, Romero-Quintana JG, Ramos-Payan R, Ruíz-García E, López-Camarillo C. SOX9 Stem-Cell Factor: Clinical and Functional Relevance in Cancer. JOURNAL OF ONCOLOGY 2019; 2019:6754040. [PMID: 31057614 PMCID: PMC6463569 DOI: 10.1155/2019/6754040] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/21/2019] [Indexed: 12/15/2022]
Abstract
Transcriptional and epigenetic embryonic programs can be reactivated in cancer cells. As result, a specific subset of undifferentiated cells with stem-cells properties emerges and drives tumorigenesis. Recent findings have shown that ectoderm- and endoderm-derived tissues continue expressing stem-cells related transcription factors of the SOX-family of proteins such as SOX2 and SOX9 which have been implicated in the presence of cancer stem-like cells (CSCs) in tumors. Currently, there is enough evidence suggesting an oncogenic role for SOX9 in different types of human cancers. This review provides a summary of the current knowledge about the involvement of SOX9 in development and progression of cancer. Understanding the functional roles of SOX9 and clinical relevance is crucial for developing novel treatments targeting CSCs in cancer.
Collapse
Affiliation(s)
- Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Mariana Avendaño-Félix
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Erik Lizárraga-Verdugo
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Mercedes Bermúdez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | | | - Rosalío Ramos-Payan
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Erika Ruíz-García
- Laboratorio de Medicina Traslacional y Departamento de Tumores Gastro-Intestinales, Instituto Nacional de Cancerología. CDMX, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX, Mexico
| |
Collapse
|
47
|
Ogawa Y, Terao M, Hara S, Tamano M, Okayasu H, Kato T, Takada S. Mapping of a responsible region for sex reversal upstream of Sox9 by production of mice with serial deletion in a genomic locus. Sci Rep 2018; 8:17514. [PMID: 30504911 PMCID: PMC6269501 DOI: 10.1038/s41598-018-35746-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/09/2018] [Indexed: 11/09/2022] Open
Abstract
Sox9 plays critical roles in testis formation. By mapping four familial cases of disorders of sexual development, a 32.5 kb sequence located far upstream of SOX9 was previously identified as being a commonly deleted region and named the XY sex reversal region (XYSR). To narrow down a responsible sequence in XYSR, we generated mutant mice with a series of deletions in XYSR by application of the CRISPR/Cas9 system, using a mixture of sgRNAs targeting several kilobase (kb) intervals in the region. When the whole XYSR corresponding sequence in mice was deleted in XY karyotype individuals, the mutation resulted in female offspring, suggesting that an expression mechanism of SOX9/Sox9 through XYSR is conserved in human and mouse. Male-to-female sex reversal was found in mice with a 4.8 kb deletion. We identified a sequence conserved among humans, mice, and opossum, the deletion of which (783 bp) in mice resulted in male-to-female sex reversal. The sequence includes a recently reported critical gonad enhancer for Sox9. Although it cannot be concluded that the human sequence is responsible for XYSR, it is likely. This method is applicable for fine mapping of responsible sequences for disease-causing deletions especially with regard to rare diseases.
Collapse
Affiliation(s)
- Yuya Ogawa
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Miho Terao
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Satoshi Hara
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Moe Tamano
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Haruka Okayasu
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Tomoko Kato
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan.,Tokyo Metropolitan Institute of Medical Science, Regenerative Medicine Project, Tokyo, 156-8506, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan.
| |
Collapse
|
48
|
Shimizu N, Matsuda M. Identification of a Novel Zebrafish Mutant Line that Develops Testicular Germ Cell Tumors. Zebrafish 2018; 16:15-28. [PMID: 30300574 DOI: 10.1089/zeb.2018.1604] [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] [Indexed: 11/12/2022] Open
Abstract
Testicular tumors are the most common solid malignant tumors in men 20-35 years of age. Although most of testicular tumors are curable, current treatments still fail in 15%-20% of patients. However, insufficient understanding of the molecular basis and lack of animal models limit development of more effective treatments. This study reports the identification of a novel zebrafish mutant line, ns1402, which develops testicular germ cell tumors (TGCTs). While both male and female ns1402 mutants were fertile at young age, male ns1402 mutants became infertile as early as 9 months of age. This infertility was associated with progressive loss of mature sperm. Failure of spermatogenesis was, at least in part, explained by progressive loss of mature Leydig cells, a source of testosterone that is essential for spermatogenesis. Interestingly, TGCTs in ns1402 mutants contained a large number of Sertoli cells and gene expression profiles of Sertoli cells were altered before loss of mature Leydig cells. This suggests that changes in Sertoli cell properties happened first, followed by loss of mature Leydig cells and failure of spermatogenesis. Taken together, this study emphasizes the importance of cell-cell interactions and cell signaling in the testis for spermatogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Miho Matsuda
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
49
|
Abstract
Who is the determining factor for the sex of the offspring—mother, father, or both parents? This fundamental hypothesis proposes a new model of sex determination, challenging the existing dogma that the male Y chromosome of the father is the sole determinant of the sex of the offspring. According to modern science, the 3 X chromosomes (male XY and female XX) are assumed to be similar, and the sex of the offspring is determined after the zygote is formed. In contrast to this, the new hypothesis based on theoretical research proposes that the 3 X chromosomes can be differentiated, based on the presence of Barr bodies. The first X in female XX chromosomes and X in male XY chromosomes are similar as they lack Barr body and are hereby denoted as ‘X’ and referred to as ancestral chromosomes. The second X chromosome in the female cells which is a Barr body, denoted as X, is different. This X chromosome along with the Y chromosome are referred to as parental chromosomes. Sperm with a Y chromosome can only fuse with an ovum containing the ‘X’ chromosome. Similarly, sperm with the ‘X’ chromosome can only fuse with an ovum containing the X chromosome. Cell biology models of gametogenesis and fertilization were simulated with the new hypothesis model and assessed. Only chromosomes that participated in recombination could unite to form the zygote. This resulted in a paradigm shift in our understanding of sex determination, as both parents were found to be equally responsible for determining the sex of the offspring. The gender of the offspring is determined during the prezygotic stage itself and is dependent on natural selection. A new dimension has been given to inheritance of chromosomes. This new model also presents a new nomenclature for pedigree charts. This work of serendipity may contribute to future research in cell biology, gender studies, genome analysis, and genetic disorders including cancer.
Collapse
|
50
|
O'Neill MJ, O'Neill RJ. Sex chromosome repeats tip the balance towards speciation. Mol Ecol 2018; 27:3783-3798. [PMID: 29624756 DOI: 10.1111/mec.14577] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/08/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
Because sex chromosomes, by definition, carry genes that determine sex, mutations that alter their structural and functional stability can have immediate consequences for the individual by reducing fertility, but also for a species by altering the sex ratio. Moreover, the sex-specific segregation patterns of heteromorphic sex chromosomes make them havens for selfish genetic elements that not only create suboptimal sex ratios but can also foster sexual antagonism. Compensatory mutations to mitigate antagonism or return sex ratios to a Fisherian optimum can create hybrid incompatibility and establish reproductive barriers leading to species divergence. The destabilizing influence of these selfish elements is often manifest within populations as copy number variants (CNVs) in satellite repeats and transposable elements (TE) or as CNVs involving sex-determining genes, or genes essential to fertility and sex chromosome dosage compensation. This review catalogs several examples of well-studied sex chromosome CNVs in Drosophilids and mammals that underlie instances of meiotic drive, hybrid incompatibility and disruptions to sex differentiation and sex chromosome dosage compensation. While it is difficult to pinpoint a direct cause/effect relationship between these sex chromosome CNVs and speciation, it is easy to see how their effects in creating imbalances between the sexes, and the compensatory mutations to restore balance, can lead to lineage splitting and species formation.
Collapse
Affiliation(s)
- Michael J O'Neill
- Institute for Systems Genomics and Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Rachel J O'Neill
- Institute for Systems Genomics and Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| |
Collapse
|