1
|
Estermann MA, Grimm S, Kitakule A, Rodriguez K, Brown P, McClelland K, Amato C, Yao HHC. NR2F2 regulation of interstitial to fetal Leydig cell differentiation in the testis: insights into differences of sex development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613312. [PMID: 39345510 PMCID: PMC11429913 DOI: 10.1101/2024.09.16.613312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Testicular fetal Leydig cells are a specialized cell type responsible for embryo masculinization. Fetal Leydig cells produce androgens, that induce the differentiation of male reproductive system and sexual characteristics. Deficiencies in Leydig cell differentiation leads to various disorders of sex development and male reproductive defects such as ambiguous genitalia, hypospadias, cryptorchidism, and infertility. Fetal Leydig cells are thought to originate from proliferating progenitor cells in the testis interstitium, marked by genes like Arx , Pdgfra , Tcf21 and Wnt5a . However, the precise mechanisms governing the transition from interstitial cells to fetal Leydig cells remain elusive. Through integrated approaches involving mouse models and single-nucleus multiomic analyses, we discovered that fetal Leydig cells originate from a Nr2f2 -positive non-steroidogenic interstitial cell population. Embryonic deletion of Nr2f2 in mouse testes resulted in disorders of sex development, including dysgenic testes, Leydig cell hypoplasia, cryptorchidism, and hypospadias. We found that NR2F2 promotes the progenitor cell fate while suppresses Leydig cell differentiation by directly and indirectly controlling a cohort of transcription factors and downstream genes. Bioinformatic analyses of single-nucleus ATAC-seq and NR2F2 ChIP-seq data revealed putative transcription factors co-regulating the process of interstitial to Leydig cell differentiation. Collectively, our findings not only highlight the critical role of Nr2f2 in orchestrating the transition from interstitial cells to fetal Leydig cells, but also provide molecular insight into the disorders of sex development as a result of Nr2f2 mutations.
Collapse
|
2
|
Gao Y, Wang Z, Long Y, Yang L, Jiang Y, Ding D, Teng B, Chen M, Yuan J, Gao F. Unveiling the roles of Sertoli cells lineage differentiation in reproductive development and disorders: a review. Front Endocrinol (Lausanne) 2024; 15:1357594. [PMID: 38699384 PMCID: PMC11063913 DOI: 10.3389/fendo.2024.1357594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/07/2024] [Indexed: 05/05/2024] Open
Abstract
In mammals, gonadal somatic cell lineage differentiation determines the development of the bipotential gonad into either the ovary or testis. Sertoli cells, the only somatic cells in the spermatogenic tubules, support spermatogenesis during gonadal development. During embryonic Sertoli cell lineage differentiation, relevant genes, including WT1, GATA4, SRY, SOX9, AMH, PTGDS, SF1, and DMRT1, are expressed at specific times and in specific locations to ensure the correct differentiation of the embryo toward the male phenotype. The dysregulated development of Sertoli cells leads to gonadal malformations and male fertility disorders. Nevertheless, the molecular pathways underlying the embryonic origin of Sertoli cells remain elusive. By reviewing recent advances in research on embryonic Sertoli cell genesis and its key regulators, this review provides novel insights into sex determination in male mammals as well as the molecular mechanisms underlying the genealogical differentiation of Sertoli cells in the male reproductive ridge.
Collapse
Affiliation(s)
- Yang Gao
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zican Wang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Yue Long
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lici Yang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongjian Jiang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Dongyu Ding
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Baojian Teng
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
3
|
Shon WJ, Seong H, Song JW, Shin DM. Taste receptor type 1 member 3 is required for the fertility of male mice. Heliyon 2024; 10:e24577. [PMID: 38312691 PMCID: PMC10835302 DOI: 10.1016/j.heliyon.2024.e24577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Male infertility is a global health concern. However, its underlying pathophysiology remains unclear. Taste receptor type 1 member 3 (TAS1R3) is highly expressed in the testes, indicating its potential involvement in male fertility. Using wild-type and Tas1r3 knockout (KO) mice, we investigated whether TAS1R3 modulates male reproductive function. Tas1r3 KO mice exhibited reduced male fertility compared to WT mice, with fewer live pups per litter and a delayed first litter. Testicular transcriptome analysis indicated suppressed PKA/CREB/StAR signaling-mediated testosterone synthesis in Tas1r3 KO mice. In silico single-cell RNA sequencing revealed considerably higher Tas1r3 expression in Leydig cells than in other testicular cell subtypes. An in vitro study validated that Tas1r3 knockdown downregulated the expression of Creb1 and steroidogenic genes in Leydig cells. Our results suggest that testicular TAS1R3 is intricately involved in male reproduction via the PKA/CREB/StAR signaling pathway, highlighting its potential as a promising target for addressing male infertility.
Collapse
Affiliation(s)
- Woo-Jeong Shon
- Research Institute of Human Ecology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Hobin Seong
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Jae Won Song
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dong-Mi Shin
- Research Institute of Human Ecology, Seoul National University, Gwanak-gu, Seoul 08826, Republic of Korea
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-gu, Seoul 08826, Republic of Korea
| |
Collapse
|
4
|
Ruthig VA, Hatkevich T, Hardy J, Friedersdorf MB, Mayère C, Nef S, Keene JD, Capel B. The RNA binding protein DND1 is elevated in a subpopulation of pro-spermatogonia and targets chromatin modifiers and translational machinery during late gestation. PLoS Genet 2023; 19:e1010656. [PMID: 36857387 PMCID: PMC10010562 DOI: 10.1371/journal.pgen.1010656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 03/13/2023] [Accepted: 02/06/2023] [Indexed: 03/02/2023] Open
Abstract
DND1 is essential to maintain germ cell identity. Loss of Dnd1 function results in germ cell differentiation to teratomas in some inbred strains of mice or to somatic fates in zebrafish. Using our knock-in mouse line in which a functional fusion protein between DND1 and GFP is expressed from the endogenous locus (Dnd1GFP), we distinguished two male germ cell (MGC) populations during late gestation cell cycle arrest (G0), consistent with recent reports of heterogeneity among MGCs. Most MGCs express lower levels of DND1-GFP (DND1-GFP-lo), but some MGCs express elevated levels of DND1-GFP (DND1-GFP-hi). A RNA-seq time course confirmed high Dnd1 transcript levels in DND1-GFP-hi cells along with 5-10-fold higher levels for multiple epigenetic regulators. Using antibodies against DND1-GFP for RNA immunoprecipitation (RIP)-sequencing, we identified multiple epigenetic and translational regulators that are binding targets of DND1 during G0 including DNA methyltransferases (Dnmts), histone deacetylases (Hdacs), Tudor domain proteins (Tdrds), actin dependent regulators (Smarcs), and a group of ribosomal and Golgi proteins. These data suggest that in DND1-GFP-hi cells, DND1 hosts coordinating mRNA regulons that consist of functionally related and localized groups of epigenetic enzymes and translational components.
Collapse
Affiliation(s)
- Victor A. Ruthig
- Sexual Medicine Lab, Department of Urology, Weill Cornell Medicine, New York, New York, United States of America
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Talia Hatkevich
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Josiah Hardy
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew B. Friedersdorf
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Geneva, Switzerland
| | - Jack D. Keene
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
5
|
Bhattacharya I, Dey S. Emerging concepts on Leydig cell development in fetal and adult testis. Front Endocrinol (Lausanne) 2023; 13:1086276. [PMID: 36686449 PMCID: PMC9851038 DOI: 10.3389/fendo.2022.1086276] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Leydig cells (Lc) reside in the interstitial compartment of the testis and are the target of Luteinising hormone (LH) for Testosterone (T) production, thus critically regulates male fertility. Classical histological studies have identified two morphologically different populations of Lc during testicular development [fetal (FLc) and adult (ALc)]. Recent progress in ex vivo cell/organ culture, genome-wide analysis, genetically manipulated mouse models, lineage tracing, and single-cell RNA-seq experiments have revealed the diverse cellular origins with differential transcriptomic and distinct steroidogenic outputs of these populations. FLc originates from both coelomic epithelium and notch-active Nestin-positive perivascular cells located at the gonad-mesonephros borders, and get specified as Nr5a1 (previously known as Ad4BP/SF-1) expressing cells by embryonic age (E) 12.5 days in fetal mouse testes. These cells produce androstenedione (precursor of T, due to lack of HSD17β3 enzyme) and play critical a role in initial virilization and patterning of the male external genitalia. However, in neonatal testis, FLc undergoes massive regression/dedifferentiation and gradually gets replaced by T-producing ALc. Very recent studies suggest a small fraction (5-20%) of FLc still persists in adult testis. Both Nestin-positive perivascular cells and FLc are considered to be the progenitor populations for ALc. This minireview article summarizes the current understanding of Lc development in fetal and adult testes highlighting their common or diverse cellular (progenitor/stem) origins with respective functional significance in both rodents and primates. (227 words).
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Periye, Kerala, India
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
6
|
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: 5.0] [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
|
7
|
Cunha GR, Cao M, Aksel S, Derpinghaus A, Baskin LS. Mouse-human species differences in early testicular development and its implications. Differentiation 2023; 129:79-95. [PMID: 35667976 DOI: 10.1016/j.diff.2022.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/16/2022] [Accepted: 04/24/2022] [Indexed: 01/25/2023]
Abstract
The mouse has been used as a model of human organogenesis with the tacit assumption that morphogenetic and molecular mechanisms in mice are translatable to human organogenesis. While many morphogenetic and molecular mechanisms are shared in mice and humans, many anatomic, morphogenetic, and molecular differences have been noted. Two critical gaps in our knowledge prevent meaningful comparisons of mouse versus human testicular development: (a) human testicular development is profoundly under-represented in the literature, and (b) an absence of a detailed day-by-day ontogeny of mouse testicular development from E11.5 to E16.5 encompassing the ambisexual stage to seminiferous cord formation. To address these deficiencies, histologic and immunohistochemical studies were pursued in comparable stages of mouse and human testicular development with a particular emphasis on Leydig, Sertoli and myoid cells through review of the literature and new observations. For example, an androgen-receptor-positive testicular medulla is present in the developing human testis but not in the developing mouse testis. The human testicular medulla and associated mesonephros were historically described as the source of Sertoli cells in seminiferous cords. Consistent with this idea, the profoundly androgen receptor (AR)-positive human testicular medulla was shown to be a zone of mesenchymal to epithelial transition and a zone from which AR-positive cells appear to migrate into the human testicular cortex. While mouse Sertoli and Leydig cells have been proposed to arise from coelomic epithelium, Sertoli (SOX9) or Leydig (HSD3B1) cell markers are absent from the immediate coelomic zone of the developing human testis, perhaps because Leydig and Sertoli cell precursors are undifferentiated when they egress from the coelomic epithelium. The origin of mouse and human myoid cells remains unclear. This study provides a detailed comparison of the early stages of testicular development in human and mouse emphasizing differences in developmental processes.
Collapse
Affiliation(s)
- Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
8
|
Shao J, Wang J, Wen X, Xie J, Huang F, Guan X, Hao X, Duan P, Chen C, Chen H. Effects of aging and macrophages on mice stem Leydig cell proliferation and differentiation in vitro. Front Endocrinol (Lausanne) 2023; 14:1139281. [PMID: 37051204 PMCID: PMC10083278 DOI: 10.3389/fendo.2023.1139281] [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: 01/06/2023] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Testosterone plays a critical role in maintaining reproductive functions and well-beings of the males. Adult testicular Leydig cells (LCs) produce testosterone and are generated from stem Leydig cells (SLCs) during puberty through adulthood. In addition, macrophages are critical in the SLC regulatory niche for normal testicular function. Age-related reduction in serum testosterone contributes to a number of metabolic and quality-of-life changes in males, as well as age-related changes in immunological functions. How aging and testicular macrophages may affect SLC function is still unclear. METHODS SLCs and macrophages were purified from adult and aged mice via FACS using CD51 as a marker protein. The sorted cells were first characterized and then co-cultured in vitro to examine how aging and macrophages may affect SLC proliferation and differentiation. To elucidate specific aging effects on both cell types, co-culture of sorted SLCs and macrophages were also carried out across two ages. RESULTS CD51+ (weakly positive) and CD51++ (strongly positive) cells expressed typical SLC and macrophage markers, respectively. However, with aging, both cell types increased expression of multiple cytokine genes, such as IL-1b, IL-6 and IL-8. Moreover, old CD51+ SLCs reduced their proliferation and differentiation, with a more significant reduction in differentiation (2X) than proliferation (30%). Age matched CD51++ macrophages inhibited CD51+ SLC development, with a more significant reduction in old cells (60%) than young (40%). Crossed-age co-culture experiments indicated that the age of CD51+ SLCs plays a more significant role in determining age-related inhibitory effects. In LC lineage formation, CD51+ SLC had both reduced LC lineage markers and increased myoid cell lineage markers, suggesting an age-related lineage shift for SLCs. CONCLUSION The results suggest that aging affected both SLC function and their regulatory niche cell, macrophages.
Collapse
Affiliation(s)
- Jingjing Shao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou City, Department of Pediatric Urology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiexia Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xin Wen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiajia Xie
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fu Huang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoju Guan
- Key Laboratory of Children Genitourinary Diseases of Wenzhou City, Department of Pediatric Urology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinrui Hao
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ping Duan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Haolin Chen, ; Congde Chen, ; Ping Duan,
| | - Congde Chen
- Key Laboratory of Children Genitourinary Diseases of Wenzhou City, Department of Pediatric Urology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Haolin Chen, ; Congde Chen, ; Ping Duan,
| | - Haolin Chen
- Zhejiang Provincial Key Laboratory of Anesthesiology, Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Children Genitourinary Diseases of Wenzhou City, Department of Pediatric Urology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pharmacology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Haolin Chen, ; Congde Chen, ; Ping Duan,
| |
Collapse
|
9
|
Inoue M, Baba T, Takahashi F, Terao M, Yanai S, Shima Y, Saito D, Sugihara K, Miura T, Takada S, Suyama M, Ohkawa Y, Morohashi KI. Tmsb10 triggers fetal Leydig differentiation by suppressing the RAS/ERK pathway. Commun Biol 2022; 5:974. [PMID: 36109592 PMCID: PMC9478096 DOI: 10.1038/s42003-022-03941-5] [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: 03/18/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
Leydig cells in fetal testes play crucial roles in masculinizing fetuses through androgen production. Gene knockout studies have revealed that growth factors are implicated in fetal Leydig cell (FLC) differentiation, but little is known about the mechanisms regulating this process. We investigate this issue by characterizing FLC progenitor cells using single-cell RNA sequencing. The sequence datasets suggest that thymosin β10 (Tmsb10) is transiently upregulated in the progenitors. While studying the function of Tmsb10, we reveal that platelet-derived growth factor (PDGF) regulates ciliogenesis through the RAS/ERK and PI3K/AKT pathways, and thereby promotes desert hedgehog (DHH)-dependent FLC differentiation. Tmsb10 expressed in the progenitor cells induces their differentiation into FLCs by suppressing the RAS/ERK pathway. Through characterizing the transiently expressed Tmsb10 in the FLC progenitors, this study unveils the molecular process of FLC differentiation and shows that it is cooperatively induced by DHH and PDGF. Investigation of fetal Leydig progenitors shows that thymosin β10 (Tmsb10) suppresses the RAS/ERK pathway, inducing progenitor differentiation into fetal Leydig cells.
Collapse
|
10
|
Lustofin S, Kamińska A, Brzoskwinia M, Cyran J, Kotula-Balak M, Bilińska B, Hejmej A. Nuclear and Membrane Receptors for Sex Steroids Are Involved in the Regulation of Delta/Serrate/LAG-2 Proteins in Rodent Sertoli Cells. Int J Mol Sci 2022; 23:ijms23042284. [PMID: 35216398 PMCID: PMC8876387 DOI: 10.3390/ijms23042284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Delta/Serrate/LAG-2 (DSL) proteins, which serve as ligands for Notch receptors, mediate direct cell–cell interactions involved in the determination of cell fate and functioning. The present study aimed to explore the role of androgens and estrogens, and their receptors in the regulation of DSL proteins in Sertoli cells. To this end, primary rat Sertoli cells and TM4 Sertoli cell line were treated with either testosterone or 17β-estradiol and antagonists of their receptors. To confirm the role of particular receptors, knockdown experiments were performed. mRNA and protein expressions of Jagged1 (JAG1), Delta-like1 (DLL1), and Delta-like4 (DLL4) were analyzed using RT-qPCR, Western blot, and immunofluorescence. Testosterone caused downregulation of JAG1 and DLL1 expression, acting through membrane androgen receptor ZRT- and Irt-like protein 9 (ZIP9) or nuclear androgen receptor (AR), respectively. DLL4 was stimulated by testosterone in the manner independent of AR and ZIP9 in Sertoli cells. The expression of all studied DSL proteins was upregulated by 17β-estradiol. Estrogen action on JAG1 and DLL1 was mediated chiefly via estrogen receptor α (ERα), while DLL4 was controlled via estrogen receptor β (ERβ) and membrane G-protein-coupled estrogen receptor (GPER). To summarize, the co-operation of nuclear and membrane receptors for sex steroids controls DSL proteins in Sertoli cells, contributing to balanced Notch signaling activity in seminiferous epithelium.
Collapse
Affiliation(s)
- Sylwia Lustofin
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
| | - Alicja Kamińska
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
| | - Małgorzata Brzoskwinia
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
| | - Joanna Cyran
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
| | - Małgorzata Kotula-Balak
- Department of Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, 30-059 Krakow, Poland;
| | - Barbara Bilińska
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
| | - Anna Hejmej
- Department of EndocrinologyInstitute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, 30-387 Krakow, Poland; (S.L.); (A.K.); (M.B.); (J.C.); (B.B.)
- Correspondence:
| |
Collapse
|
11
|
Peroxisome Proliferator-Activated Receptor γ, but Not α or G-Protein Coupled Estrogen Receptor Drives Functioning of Postnatal Boar Testis-Next Generation Sequencing Analysis. Animals (Basel) 2021; 11:ani11102868. [PMID: 34679887 PMCID: PMC8532933 DOI: 10.3390/ani11102868] [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: 08/09/2021] [Revised: 09/19/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary As of now, the Next Generation Sequencing (NGS) analysis has not been utilized to identify biological processes and signaling pathways that are regulated in the boar postnatal testes. Our prior studies revealed that the peroxisome proliferator-activated receptor (PPAR) and G-protein coupled estrogen receptor (GPER) were significant for the morpho-functional status of testicular cells. Here, the pharmacological blockage of PPARα, PPARγ or GPER was performed in ex vivo immature boar testes. The NGS results showed 382 transcripts with an altered expression. The blockage by the PPARγ antagonist markedly affected biological processes such as: drug metabolism (genes: Ctsh, Duox2, Atp1b1, Acss2, Pkd2, Aldh2, Hbb, Sdhd, Cox3, Nd4, Nd5, Cytb, Cbr1, and Pid1), adhesion (genes: Plpp3, Anxa1, Atp1b1, S100a8, Cd93, Ephb4, Vsir, Cldn11, Gpc4, Fermt3, Dusp26, Sox9, and Cdh5) and tube development (genes: Ctsh, Mmp14, Dll4, Anxa1, Ephb4, Pkd2, Angptl4, Robo4, Sox9, Hikeshi, Ing2, Loc100738836, and Rarres2), as well as the Notch signaling pathway. This was not the case for the PPARα or GPER antagonists. Our observations suggested that PPARγ may be the principal player in the management of the development and function of boar testes during the early postnatal window. Moreover, due to a highly similar porcine gene expression pattern to human homologues genes, our results can be used to understand both animal and human testes physiology and to predict or treat pathological processes. Abstract Porcine tissue gene expression is highly similar to the expression of homologous genes in humans. Based on this fact, the studies on porcine tissues can be employed to understand human physiology and to predict or treat diseases. Our prior studies clearly showed that there was a regulatory partnership of the peroxisome proliferator-activated receptor (PPAR) and the G-protein coupled membrane estrogen receptor (GPER) that relied upon the tumorigenesis of human and mouse testicular interstitial cells, as well as the PPAR-estrogen related receptor and GPER–xenoestrogen relationships which affected the functional status of immature boar testes. The main objective of this study was to identify the biological processes and signaling pathways governed by PPARα, PPARγ and GPER in the immature testes of seven-day-old boars after pharmacological receptor ligand treatment. Boar testicular tissues were cultured in an organotypic system with the respective PPARα, PPARγ or GPER antagonists. To evaluate the effect of the individual receptor deprivation in testicular tissue on global gene expression, Next Generation Sequencing was performed. Bioinformatic analysis revealed 382 transcripts with altered expression. While tissues treated with PPARα or GPER antagonists showed little significance in the enrichment analysis, the antagonists challenged with the PPARγ antagonist displayed significant alterations in biological processes such as: drug metabolism, adhesion and tubule development. Diverse disruption in the Notch signaling pathway was also observed. The findings of our study proposed that neither PPARα nor GPER, but PPARγ alone seemed to be the main player in the regulation of boar testes functioning during early the postnatal developmental window.
Collapse
|
12
|
Kumar S, Kim HJ, Lee CH, Choi HS, Lee K. Leydig Cell-Specific DAX1-Deleted Mice Has Higher Testosterone Level in the Testis During Pubertal Development. Reprod Sci 2021; 29:955-962. [PMID: 33891289 DOI: 10.1007/s43032-021-00554-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/22/2021] [Indexed: 11/28/2022]
Abstract
Testosterone, the male sex hormone, is necessary for the development and function of the male reproductive system. Biosynthesis of testosterone in mammals mainly occurs in testicular Leydig cells. Many proteins such as P450c17, 3β-HSD, and StAR are involved in testicular steroidogenesis. DAX1 is essential for sex development and interacts with nuclear receptors such as steroidogenic factor 1 to inhibit steroidogenesis. In this study, we investigated the role of DAX1 in testicular steroidogenesis in vivo by generating Leydig cell-specific DAX1-knockout mice. Radioimmunoassay revealed that the levels of testosterone and progesterone were higher in Leydig cell-specific DAX1-knockout testes than in the testes from wild-type mice during the first 3-4 weeks of aging. In addition, the expression levels of steroidogenic genes, such as StAR, P450c17, P450scc, and 3β-HSD, were considerably higher in the testes from DAX1-knockout mice. DAX1-deficient mouse testes seemed to attain early puberty with the acceleration of germ cell development. These data suggest that DAX1 regulates the expression of steroidogenic genes, and thereby controls and fine-tunes steroidogenesis during testis development.
Collapse
Affiliation(s)
- Sudeep Kumar
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Hyo Jeong Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Keesook Lee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
13
|
Ruthig VA, Yokonishi T, Friedersdorf MB, Batchvarova S, Hardy J, Garness JA, Keene JD, Capel B. A transgenic DND1GFP fusion allele reports in vivo expression and RNA-binding targets in undifferentiated mouse germ cells†. Biol Reprod 2021; 104:861-874. [PMID: 33394034 PMCID: PMC8324984 DOI: 10.1093/biolre/ioaa233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/23/2020] [Accepted: 12/30/2020] [Indexed: 01/20/2023] Open
Abstract
In vertebrates, the RNA-binding protein (RBP) dead end 1 (DND1) is essential for primordial germ cell (PGC) survival and maintenance of cell identity. In multiple species, Dnd1 loss or mutation leads to severe PGC loss soon after specification or, in some species, germ cell transformation to somatic lineages. Our investigations into the role of DND1 in PGC specification and differentiation have been limited by the absence of an available antibody. To address this problem, we used CRISPR/Cas9 gene editing to establish a transgenic mouse line carrying a DND1GFP fusion allele. We present imaging analysis of DND1GFP expression showing that DND1GFP expression is heterogeneous among male germ cells (MGCs) and female germ cells (FGCs). DND1GFP was detected in MGCs throughout fetal life but lost from FGCs at meiotic entry. In postnatal and adult testes, DND1GFP expression correlated with classic markers for the premeiotic spermatogonial population. Utilizing the GFP tag for RNA immunoprecipitation (RIP) analysis in MGCs validated this transgenic as a tool for identifying in vivo transcript targets of DND1. The DND1GFP mouse line is a novel tool for isolation and analysis of embryonic and fetal germ cells, and the spermatogonial population of the postnatal and adult testis.
Collapse
Affiliation(s)
- Victor A Ruthig
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | | | - Matthew B Friedersdorf
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Sofia Batchvarova
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Josiah Hardy
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Jason A Garness
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Jack D Keene
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
14
|
Gu X, Li SY, DeFalco T. Immune and vascular contributions to organogenesis of the testis and ovary. FEBS J 2021; 289:2386-2408. [PMID: 33774913 PMCID: PMC8476657 DOI: 10.1111/febs.15848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/07/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Gonad development is a highly regulated process that coordinates cell specification and morphogenesis to produce sex-specific organ structures that are required for fertility, such as testicular seminiferous tubules and ovarian follicles. While sex determination occurs within specialized gonadal supporting cells, sexual differentiation is evident throughout the entire organ, including within the interstitial compartment, which contains immune cells and vasculature. While immune and vascular cells have been traditionally appreciated for their supporting roles during tissue growth and homeostasis, an increasing body of evidence supports the idea that these cell types are critical drivers of sexually dimorphic morphogenesis of the gonad. Myeloid immune cells, such as macrophages, are essential for multiple aspects of gonadogenesis and fertility, including for forming and maintaining gonadal vasculature in both sexes at varying stages of life. While vasculature is long known for supporting organ growth and serving as an export mechanism for gonadal sex steroids in utero, it is also an important component of fetal testicular morphogenesis and differentiation; additionally, it is vital for ovarian corpus luteal function and maintenance of pregnancy. These findings point toward a new paradigm in which immune cells and blood vessels are integral components of sexual differentiation and organogenesis. In this review, we discuss the state of the field regarding the diverse roles of immune and vascular cells during organogenesis of the testis and ovary and highlight outstanding questions in the field that could stimulate new research into these previously underappreciated constituents of the gonad.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
15
|
Yang G, He Y, Yang H. The involvement of bioactive factors in the self-renewal and stemness maintenance of spermatogonial stem cells. Mol Cell Biochem 2021; 476:1813-1823. [PMID: 33459979 DOI: 10.1007/s11010-020-04028-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
Spermatogenesis is usually accompanied throughout mammalian lifetime, transmitting genetic information to the next generation, which is mainly dependent on the self-renewal and differentiation of spermatogonial stem cells (SSCs). With further investigation on profiles of SSCs, the previous prevailing orthodoxy that SSCs are unipotent stem cells to differentiate into spermatids only, has been challenged. More notably, accumulating evidence has demonstrated that SSCs are capable of giving rise to cell lineages of the three germ layers, highlighting potential important applications of SSCs for regenerative medicine. Nevertheless, it is unknown how the proliferation and stemness maintenance of SSCs are regulated intrinsically and strictly controlled in a special niche microenvironment in the seminiferous tubules. Based on the special niche microenvironment for SSCs, it is of vital interest to summarize the recent knowledge regarding several critical bioactive molecules in the self-renewal and stemness maintenance of SSCs. In this review, we discuss most recent findings about these critical bioactive factors and further address the new advances on the self-renewal and stemness maintenance of SSCs.
Collapse
Affiliation(s)
- Guoqing Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Yuqing He
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Hao Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, Shaanxi, China. .,School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, China. .,Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China.
| |
Collapse
|
16
|
Jagged1 intracellular domain modulates steroidogenesis in testicular Leydig cells. PLoS One 2020; 15:e0244553. [PMID: 33378407 PMCID: PMC7773251 DOI: 10.1371/journal.pone.0244553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022] Open
Abstract
Leydig cells represent the steroidogenic lineage of mammalian testis, which produces testosterone. Genetic evidence indicates the requirement of Notch signaling in maintaining a balance between differentiated Leydig cells and their progenitors during fetal development. In primary Leydig cells, Notch1 expression decreases with testicular development, while the expression of its ligand, Jagged1, remains relatively unchanged, suggesting that the roles of Jagged1 extend beyond Notch signaling. In addition, Jagged1 is known to be processed into its intracellular domain, which then translocate to the nucleus. In this study, we investigated the effect of Jagged1 intracellular domain (JICD) on steroidogenesis in Leydig cells. The independent overexpression of JICD in MA-10 Leydig cells was found to inhibit the activity of cAMP-induced Nur77 promoter. In addition, JICD suppressed Nur77 transactivation of the promoter of steroidogenic genes such as P450scc, P450c17, StAR, and 3β-HSD. Further, adenovirus-mediated overexpression of JICD in primary Leydig cells repressed the expression of steroidogenic genes, consequently lowering testosterone production. These results collectively suggest that steroidogenesis in testicular Leydig cells, which is regulated by LH/cAMP signaling, is fine-tuned by Jagged1 during testis development.
Collapse
|
17
|
Kamińska A, Marek S, Pardyak L, Brzoskwinia M, Bilinska B, Hejmej A. Crosstalk between Androgen-ZIP9 Signaling and Notch Pathway in Rodent Sertoli Cells. Int J Mol Sci 2020; 21:ijms21218275. [PMID: 33167316 PMCID: PMC7663815 DOI: 10.3390/ijms21218275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Our recent study demonstrated altered expression of Notch ligands, receptors, and effector genes in testes of pubertal rats following reduced androgen production or signaling. Herein we aimed to explore the role of nuclear androgen receptor (AR) and membrane androgen receptor (Zrt- and Irt-like protein 9; ZIP9) in the regulation of Notch pathway activation in rodent Sertoli cells. Experiments were performed using TM4 and 15P-1 Sertoli cell lines and rat primary Sertoli cells (PSC). We found that testosterone (10-8 M-10-6 M) increased the expression of Notch1 receptor, its active form Notch1 intracellular domain (N1ICD) (p < 0.05, p < 0.01, p < 0.001), and the effector genes Hey1 (p < 0.05, p < 0.01, p < 0.001) and Hes1 (p < 0.05, p < 0.001) in Sertoli cells. Knockdown of AR or ZIP9 as well as antiandrogen exposure experiments revealed that (i) action of androgens via both AR and ZIP9 controls Notch1/N1ICD expression and transcriptional activity of recombination signal binding protein (RBP-J), (ii) AR-dependent signaling regulates Hey1 expression, (iii) ZIP9-dependent pathway regulates Hes1 expression. Our findings indicate a crosstalk between androgen and Notch signaling in Sertoli cells and point to cooperation of classical and non-classical androgen signaling pathways in controlling Sertoli cell function.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
| | - Sylwia Marek
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
| | - Laura Pardyak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, 30-248 Kraków, Poland
| | - Małgorzata Brzoskwinia
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
| | - Barbara Bilinska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland; (A.K.); (S.M.); (L.P.); (M.B.); (B.B.)
- Correspondence:
| |
Collapse
|
18
|
Wu K, Li Y, Pan P, Li Z, Yu Y, Huang J, Ma F, Tian L, Fang Y, Wang Y, Lin H, Ge RS. Gestational vinclozolin exposure suppresses fetal testis development in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 203:111053. [PMID: 32888615 DOI: 10.1016/j.ecoenv.2020.111053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 06/11/2023]
Abstract
Vinclozolin is a common dicarboximide fungicide used to protect crops from diseases. It is also an endocrine disruptor and is thought to be related to abnormalities of the reproductive tract. However, its mechanism of inducing abnormalities of the male reproductive tract is still unclear. The purpose of this study was to study the effect of gestational vinclozolin exposure on the development of rat fetal Leydig cells. Female pregnant Sprague-Dawley rats were exposed to vinclozolin (0, 25, 50, and 100 mg/kg body weight/day) by gavage from gestational day 14-21. Vinclozolin dose-dependently reduced serum testosterone levels at doses of 50 and 100 mg/kg and the anogenital distance at 100 mg/kg. RNA-seq, qPCR, and Western blotting showed that vinclozolin down-regulated the expression of Nr5a1, Sox9, Lhcgr, Cyp11a1, Hsd3b1, Hsd17b3, Amh, Pdgfa, and Dhh and their encoded proteins. Vinclozolin reduced the number of NR2F2-positive stem Leydig cells at a dose of 100 mg/kg and enhanced autophagy in the testes. In conclusion, vinclozolin disrupts reproductive tract development and testis development in male fetal rats via several pathways.
Collapse
Affiliation(s)
- Keyang Wu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Yang Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Peipei Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Zengqiang Li
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Yige Yu
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Jianjian Huang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Feifei Ma
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Lili Tian
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Yinghui Fang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Yiyan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Han Lin
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China.
| | - Ren-Shan Ge
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
19
|
Wulandari E, I'tishom R, Sudjarwo SA. Therapy Effect of Red Dragon Fruit (Hylocereus polyrhizus) Peel Extract to Increase the Number of Sertoli Cells On BALB/c Mice (Mus musculus) Exposed to Lead Acetate. FOLIA MEDICA INDONESIANA 2020. [DOI: 10.20473/fmi.v56i2.21228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lead is the free radicals and heavy metals of major pollutants in the environment. Lead is toxic and cumulative. Red dragon fruit (Hylocereus polyrhizus) peel extract is a natural antioxidant. Red dragon fruit peel extract can be used to stabilize free radicals by supplementing electron deficiencies and inhibiting chain reactions. This study aimed to analyze therapy effect of red dragon fruit peel extract to increase the number of Sertoli cells on BALB/c mice (Mus musculus) exposed to lead acetate. Fourty mice were divided into 5 groups (each group consisted of 8 mice). K- group was the control group without lead acetate and dragon fruit peel extract administration, K + group was given 100 mg/KgBW lead acetate orally on the 1st day until day 14th.. P1, P2, and P3 group were continued with red dragon extract orally on the 15th day until 39th day. P1 with dose 250 mg/KgBW, P2 with dose 500 mg/KgBW, and P3 with dose 1000 mg/KgBW. The result showed significant difference (P<0.05) in the number of Sertoli cells between K+ and P1, P2, P3 group. In conclusion, dose 500 mg/kgBW of red dragon fruit peel extract can be used as the most effective therapy to increase the number of Sertoli cells on mice exposed to acetate lead.
Collapse
|
20
|
Pan P, Ma F, Wu K, Yu Y, Li Y, Li Z, Chen X, Huang T, Wang Y, Ge RS. Maternal exposure to zearalenone in masculinization window affects the fetal Leydig cell development in rat male fetus. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114357. [PMID: 32229375 DOI: 10.1016/j.envpol.2020.114357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 06/10/2023]
Abstract
Zearalenone is a phenolic Fusarium mycotoxin, which is ubiquitous in human and animal feedstuff and often co-occurs with other mycotoxins. ZEA has been reported to disturb Leydig cell function and even cause the apoptosis to the Leydig cells. However, the effects of gestational exposure to zearalenone on fetal Leydig cells and the underlying mechanism remain unknown. Sprague Dawley dams were daily gavaged with 0, 2.5, 5, 10, and 20 mg/kg body weight ZEA from gestational day 14-21. On gestational day 21, rats were euthanized and serum testosterone levels were measured, and testes were collected for further evaluation of Leydig cell number, cell size, gene, and protein expression. Zearalenone significantly decreased anogenital distance and its index of male fetus, serum testosterone levels, Leydig cell proteins (SCARB1, STAR, CYP11A1, CYP17A1, and INSL3), and fetal Leydig cell number at 10 and/or 20 mg/kg by delaying the commitment of stem Leydig cells into the Leydig cell lineage and proliferation. Further study found that Notch signaling (RFNG, PSEN1, NOTCH1, and NOTCH3) was up-regulated by zearalenone. In conclusion, gestational exposure to high doses of zearalenone (10 and 20 mg/kg) blocks fetal Leydig cell development, thus possibly causing the anomalies of the male reproductive tract.
Collapse
Affiliation(s)
- Peipei Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Feifei Ma
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Keyang Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yige Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yang Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Zengqiang Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xiuxiu Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Tongliang Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yiyan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
21
|
Heinrich A, DeFalco T. Essential roles of interstitial cells in testicular development and function. Andrology 2020; 8:903-914. [PMID: 31444950 PMCID: PMC7036326 DOI: 10.1111/andr.12703] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Testicular architecture and sperm production are supported by a complex network of communication between various cell types. These signals ensure fertility by: regulating spermatogonial stem/progenitor cells; promoting steroidogenesis; and driving male-specific differentiation of the gonad. Sertoli cells have long been assumed to be the major cellular player in testis organogenesis and spermatogenesis. However, cells in the interstitial compartment, such as Leydig, vascular, immune, and peritubular cells, also play prominent roles in the testis but are less well understood. OBJECTIVES Here, we aim to outline our current knowledge of the cellular and molecular mechanisms by which interstitial cell types contribute to spermatogenesis and testicular development, and how these diverse constituents of the testis play essential roles in ensuring male sexual differentiation and fertility. METHODS We surveyed scientific literature and summarized findings in the field that address how interstitial cells interact with other interstitial cell populations and seminiferous tubules (i.e., Sertoli and germ cells) to support spermatogenesis, male-specific differentiation, and testicular function. These studies focused on 4 major cell types: Leydig cells, vascular cells, immune cells, and peritubular cells. RESULTS AND DISCUSSION A growing number of studies have demonstrated that interstitial cells play a wide range of functions in the fetal and adult testis. Leydig cells, through secretion of hormones and growth factors, are responsible for steroidogenesis and progression of spermatogenesis. Vascular, immune, and peritubular cells, apart from their traditionally acknowledged physiological roles, have a broader importance than previously appreciated and are emerging as essential players in stem/progenitor cell biology. CONCLUSION Interstitial cells take part in complex signaling interactions with both interstitial and tubular cell populations, which are required for several biological processes, such as steroidogenesis, Sertoli cell function, spermatogenesis, and immune regulation. These various processes are essential for testicular function and demonstrate how interstitial cells are indispensable for male fertility.
Collapse
Affiliation(s)
- Anna Heinrich
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7045, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7045, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Suite E-870, Cincinnati, OH, 45267, USA
| |
Collapse
|
22
|
Kamińska A, Marek S, Pardyak L, Brzoskwinia M, Pawlicki P, Bilińska B, Hejmej A. Disruption of androgen signaling during puberty affects Notch pathway in rat seminiferous epithelium. Reprod Biol Endocrinol 2020; 18:30. [PMID: 32299422 PMCID: PMC7161021 DOI: 10.1186/s12958-020-00582-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/04/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Onset of spermatogenesis at puberty is critically dependent on the activity of hypothalamic-pituitary-gonadal axis and testosterone production by Leydig cells. The aim of this study was to examine whether activation of Notch receptors and expression of Notch ligands and effector genes in rat seminiferous epithelium are controlled by androgen signaling during puberty. METHODS Peripubertal (5-week-old) Wistar rats received injections of flutamide (50 mg/kg bw) daily for 7 days to reduce androgen receptor (AR) signaling or a single injection of ethanedimethane sulphonate (EDS; 75 mg/kg bw) to reduce testosterone production. Gene and protein expressions were analyzed by real-time RT-PCR and western blotting, respectively, protein distribution by immunohistochemistry, and steroid hormone concentrations by enzyme-linked immunosorbent assay. Statistical analyses were performed using one-way ANOVA followed by Tukey's post hoc test or by Kruskal-Wallis test, followed by Dunn's test. RESULTS In both experimental models changes of a similar nature in the expression of Notch pathway components were found. Androgen deprivation caused the reduction of mRNA and protein expression of DLL4 ligand, activated forms of Notch1 and Notch2 receptors and HES1 and HEY1 effector genes (p < 0.05, p < 0.01, p < 0.001). In contrast, DLL1, JAG1 and HES5 expressions increased in seminiferous epithelium of both flutamide and EDS-treated rats (p < 0.05, p < 0.01, p < 0.001). CONCLUSIONS Androgens and androgen receptor signaling may be considered as factors regulating Notch pathway activity and the expression of Hes and Hey genes in rat seminiferous epithelium during pubertal development. Further studies should focus on functional significance of androgen-Notch signaling cross-talk in the initiation and maintenance of spermatogenesis.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Sylwia Marek
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Laura Pardyak
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Małgorzata Brzoskwinia
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Piotr Pawlicki
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Barbara Bilińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
23
|
Capel B. WOMEN IN REPRODUCTIVE SCIENCE: To be or not to be a testis. Reproduction 2019; 158:F101-F111. [PMID: 31265995 PMCID: PMC9945370 DOI: 10.1530/rep-19-0151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/25/2019] [Indexed: 11/08/2022]
Abstract
Work that established the testis as the driver of male development, and the Y chromosome as the bearer of the male-determining gene, established a working model, and set the stage for the molecular age of mammalian sex determination. The discovery and characterization of Sry/SRY at the top of the hierarchy in mammals launched the field in two major directions. The first was to identify the downstream transcription factors and other molecular players that drive the bifurcation of Sertoli and granulosa cell differentiation. The second major direction was to understand organogenesis of the early bipotential gonad, and how divergence of its two distinct morphogenetic pathways (testis and ovary) is regulated at the cellular level. This review will summarize the early discoveries soon after Sry was identified and focus on my study of the gonad as a model of organogenesis.
Collapse
Affiliation(s)
- Blanche Capel
- 1Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
24
|
Shima Y. Development of fetal and adult Leydig cells. Reprod Med Biol 2019; 18:323-330. [PMID: 31607792 PMCID: PMC6780029 DOI: 10.1002/rmb2.12287] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/09/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In mammals, two distinct Leydig cell populations, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs), appear in the prenatal and postnatal testis, respectively. Although the functional differences between these cell types have been well described, the developmental relationship between FLCs and ALCs has not been fully understood. In this review, I focus on the cellular origins of FLCs and ALCs as well as the developmental and functional links between them. METHODS I surveyed previous reports about FLC and/or ALC development and summarized the findings. MAIN FINDINGS Fetal Leydig cells and ALCs were identified to have separate origins in the fetal and neonatal testis, respectively. However, several studies suggested that FLCs and ALCs share a common progenitor pool. Moreover, perturbation of FLC development at the fetal stage induces ALC dysfunction in adults, suggesting a functional link between FLCs and ALCs. Although the lineage relationship between FLCs and ALCs remains controversial, a recent study suggested that some FLCs dedifferentiate at the fetal stage, and that these cells serve as ALC stem cells. CONCLUSION Findings obtained from animal studies might provide clues to the causative mechanisms of male reproductive dysfunctions such as testicular dysgenesis syndrome in humans.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of AnatomyKawasaki Medical SchoolKurashikiOkayamaJapan
| |
Collapse
|
25
|
Lu E, Feng F, Wen W, Tong X, Li X, Xiao L, Li G, Wang J, Zhang C. Notch signaling inhibition induces G0/G1 arrest in murine Leydig cells. Andrologia 2019; 51:e13413. [PMID: 31523838 DOI: 10.1111/and.13413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/16/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022] Open
Abstract
As a highly evolutionarily conserved signaling pathway, Notch widely participates in cell-fate decisions and the development of various tissues and organs. In male reproduction, research on the Notch signaling pathway has mainly concentrated on germ cells and Sertoli cells. Leydig cells are the primary producers of testosterone and play important roles in spermatogenesis and maintaining secondary sexual characteristics. In this study, we used TM3 cells, a murine adult Leydig cell line, to investigate the expression profiles of Notch receptors and ligands and observe the effect of Notch signaling on the proliferation of TM3 cells. We found that Notch 1-3 and the ligands Dll-1 and Dll-4 were expressed in TM3 cells, Notch 1-3 and the ligand Dll-1 were expressed in testis interstitial Leydig cells, and Notch signaling inhibition suppressed the proliferation of TM3 cells and induced G0/G1 arrest. Inhibition of Notch signaling increased the expression of p21Waf1/Cip1 and p27. Overall, our results suggest that Notch inhibition suppresses the proliferation of TM3 cells and P21Waf1/Cip1 , and p27 may contribute to this process.
Collapse
Affiliation(s)
- Enhang Lu
- Joint Programme of Nanchang University and Queen Mary University of London, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Fen Feng
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Weihui Wen
- Department of Microbiology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Xiating Tong
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Xiang Li
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Li Xiao
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Gang Li
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Jing Wang
- Department of Microbiology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| | - Chunping Zhang
- Department of Cell Biology, Jiangxi Medicine School, Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Kamińska A, Pardyak L, Marek S, Wróbel K, Kotula-Balak M, Bilińska B, Hejmej A. Notch signaling regulates nuclear androgen receptor AR and membrane androgen receptor ZIP9 in mouse Sertoli cells. Andrology 2019; 8:457-472. [PMID: 31468707 DOI: 10.1111/andr.12691] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/24/2019] [Accepted: 07/14/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Notch signaling pathway is involved in contact-dependent communication between the cells of seminiferous epithelium, and its proper activity is important for undisturbed spermatogenesis. OBJECTIVES The aim was to assess the effect of Notch pathway inhibition on the expression of nuclear (AR) and membrane (ZIP9) androgen receptors and androgen-regulated genes, claudin-5 and claudin-11, in TM4 mouse Sertoli cell line. MATERIALS AND METHODS DAPT (γ-secretase inhibitor) treatment and recombination signal binding protein silencing were employed to reduce Notch signaling, whereas immobilized ligands were used to activate Notch pathway in TM4 cells. To reveal specific effect of each androgen receptor, AR or ZIP9 silencing was performed. RESULTS Notch pathway inhibition increased the expression of AR and ZIP9 mRNA and proteins (p < 0.01; p < 0.05) in TM4 cells, whereas incubation with Notch ligands, rDLL1 or rJAG1, reduced AR (p < 0.01; p < 0.001) and ZIP9 (p < 0.05; p < 0.01) expressions, respectively. Testosterone enhanced the expression of both receptors (p < 0.05; p < 0.01). Androgen-regulated claudin-5 and claudin-11 (p < 0.01; p < 0.001) and cAMP (p < 0.001) were elevated in Notch-inhibited cells, while activation of Notch signaling by DLL1 or JAG1 reduced claudin-11 or claudin-5 level (p < 0.01; p < 0.001), respectively. DISCUSSION Our findings indicate opposite effect of Notch and androgen signaling on the expression of androgen receptors in TM4 cells. We demonstrated that AR expression is regulated by DLL1-mediated Notch signaling, whereas JAG1 is involved in the regulation of ZIP9. The expression of both claudins and cAMP production is under inhibitory influence of Notch pathway. The effects of Notch signaling on claudin-5 and claudin-11 expression are mediated by ZIP9 and AR, respectively. CONCLUSION Notch signaling may be considered as an important pathway controlling Sertoli cell physiology, and its alterations may contribute to disturbed response of Sertoli cells to androgens.
Collapse
Affiliation(s)
- A Kamińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| | - L Pardyak
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| | - S Marek
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| | - K Wróbel
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| | - M Kotula-Balak
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland.,University Centre of Veterinary Medicine, University of Agriculture in Krakow, Krakow, Poland
| | - B Bilińska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| | - A Hejmej
- Department of Endocrinology, Faculty of Biology, Institute of Zoology & Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
27
|
Bhattacharya I, Sen Sharma S, Majumdar SS. Pubertal orchestration of hormones and testis in primates. Mol Reprod Dev 2019; 86:1505-1530. [DOI: 10.1002/mrd.23246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology & BiotechnologyHNB Garhwal University, Srinagar CampusSrinagar India
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Souvik Sen Sharma
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Subeer S. Majumdar
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
- Gene and Protein Engineering LabNational Institute of Animal BiotechnologyHyderabad India
| |
Collapse
|
28
|
Fortini F, Vieceli Dalla Sega F, Caliceti C, Lambertini E, Pannuti A, Peiffer DS, Balla C, Rizzo P. Estrogen-mediated protection against coronary heart disease: The role of the Notch pathway. J Steroid Biochem Mol Biol 2019; 189:87-100. [PMID: 30817989 DOI: 10.1016/j.jsbmb.2019.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
Abstract
Estrogen regulates a plethora of biological processes, under physiological and pathological conditions, by affecting key pathways involved in the regulation of cell proliferation, fate, survival and metabolism. The Notch receptors are mediators of communication between adjacent cells and are key determinants of cell fate during development and in postnatal life. Crosstalk between estrogen and the Notch pathway intervenes in many processes underlying the development and maintenance of the cardiovascular system. The identification of molecular mechanisms underlying the interaction between these types of endocrine and juxtacrine signaling are leading to a deeper understanding of physiological conditions regulated by these steroid hormones and, potentially, to novel therapeutic approaches to prevent pathologies linked to reduced levels of estrogen, such as coronary heart disease, and cardiotoxicity caused by hormone therapy for estrogen-receptor-positive breast cancer.
Collapse
Affiliation(s)
| | | | - Cristiana Caliceti
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Antonio Pannuti
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Daniel S Peiffer
- Oncology Research Institute, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA; Department of Microbiology and Immunology, Loyola University Chicago: Health Sciences Division, Maywood, Illinois, USA
| | - Cristina Balla
- Cardiovascular Center, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, RA, Italy; Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
29
|
Guan X, Chen F, Chen P, Zhao X, Mei H, Liu J, Lian Q, Zirkin BR, Chen H. Effects of spermatogenic cycle on Stem Leydig cell proliferation and differentiation. Mol Cell Endocrinol 2019; 481:35-43. [PMID: 30476560 PMCID: PMC6367675 DOI: 10.1016/j.mce.2018.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 11/22/2018] [Accepted: 11/22/2018] [Indexed: 01/13/2023]
Abstract
We reported previously that stem Leydig cells (SLC) on the surfaces of rat testicular seminiferous tubules are able to differentiate into Leydig cells. The proliferation and differentiation of SLCs seem likely to be regulated by niche cells, including nearby germ and Sertoli cells. Due to the cyclical nature of spermatogenesis, we hypothesized that the changes in the germ cell composition of the seminiferous tubules as spermatogenesis proceeds may affect tubule-associated SLC functions. To test this hypothesis, we compared the ability of SLCs associated with tubules at different stages of the cycle to differentiate into Leydig cells in vitro. SLCs associated with stages IX-XI were more active in proliferation and differentiation than SLCs associated with stages VII-VIII. However, when the SLCs were isolated from each of the two groups of tubules and cultured in vitro, no differences were seen in their ability to proliferate or differentiate. These results suggested that the stage-dependent local factors, not the SLCs themselves, explain the stage-dependent differences in SLC function. TGFB, produced in stage-specific fashion by Sertoli cells, is among the factors shown in previous studies to affect SLC function in vitro. When TGFB inhibitors were included in the cultures of stages IX-XI and VII-VIII tubules, stage-dependent differences in SLC development were reduced, suggesting that TGFB may be among the paracrine factors involved in the stage-dependent differences in SLC function. Taken together, the findings suggest that there is dynamic interaction between SLCs and germ/Sertoli cells within the seminiferous tubules that may affect SLC proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaojui Guan
- Department of Anesthesiology, Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Department of Gynaecology and Obstetrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fenfen Chen
- Department of Gynaecology and Obstetrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Panpan Chen
- Department of Gynaecology and Obstetrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xingxing Zhao
- Department of Anesthesiology, Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hongxia Mei
- Department of Anesthesiology, Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - June Liu
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Qingquan Lian
- Department of Anesthesiology, Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- To whom correspondence should be addressed: Haolin Chen, Ph.D., The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, 109 Western Xueyuan Road, Wenzhou, Zhejiang, 325027, China, or QingquanLian, Ph.D., Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Barry R. Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | - Haolin Chen
- Department of Anesthesiology, Perioperative Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Department of Gynaecology and Obstetrics, the Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
- To whom correspondence should be addressed: Haolin Chen, Ph.D., The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, 109 Western Xueyuan Road, Wenzhou, Zhejiang, 325027, China, or QingquanLian, Ph.D., Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
30
|
A perivascular niche for multipotent progenitors in the fetal testis. Nat Commun 2018; 9:4519. [PMID: 30375389 PMCID: PMC6207726 DOI: 10.1038/s41467-018-06996-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 10/06/2018] [Indexed: 12/27/2022] Open
Abstract
Androgens responsible for male sexual differentiation in utero are produced by Leydig cells in the fetal testicular interstitium. Leydig cells rarely proliferate and, hence, rely on constant differentiation of interstitial progenitors to increase their number during fetal development. The cellular origins of fetal Leydig progenitors and how they are maintained remain largely unknown. Here we show that Notch-active, Nestin-positive perivascular cells in the fetal testis are a multipotent progenitor population, giving rise to Leydig cells, pericytes, and smooth muscle cells. When vasculature is disrupted, perivascular progenitor cells fail to be maintained and excessive Leydig cell differentiation occurs, demonstrating that blood vessels are a critical component of the niche that maintains interstitial progenitor cells. Additionally, our data strongly supports a model in which fetal Leydig cell differentiation occurs by at least two different means, with each having unique progenitor origins and distinct requirements for Notch signaling to maintain the progenitor population. Leydig cells are steroidogenic cells in the testes and produce the androgens required for male development and spermatogenesis. Here the authors show that a multipotent progenitor population producing Leydig cells, pericytes and smooth muscle cells is maintained in a perivascular niche within the mouse fetal testis.
Collapse
|
31
|
Sun D, Dong W, Jin B, Chen G, Cai B, Deng W, Cui Y, Jin Y. Mechanisms of Yangjing Capsule in Leydig Cell Apoptosis and Testosterone Synthesis via Promoting StAR Expression. Biol Pharm Bull 2018; 41:1401-1405. [DOI: 10.1248/bpb.b18-00205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Dalin Sun
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University
| | | | - Baofang Jin
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University
| | - Guanghui Chen
- Hebei Provincial Hospital of Traditional Chinese Medicine
| | - Bin Cai
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University
| | - Weimin Deng
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University
| | - Yihan Jin
- Medical College of Qinghai University
| |
Collapse
|
32
|
Kamińska A, Pardyak L, Marek S, Górowska-Wójtowicz E, Kotula-Balak M, Bilińska B, Hejmej A. Bisphenol A and dibutyl phthalate affect the expression of juxtacrine signaling factors in rat testis. CHEMOSPHERE 2018; 199:182-190. [PMID: 29438945 DOI: 10.1016/j.chemosphere.2018.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/28/2017] [Accepted: 02/03/2018] [Indexed: 06/08/2023]
Abstract
The study was designed to examine the effects of model plastic derived compounds, bisphenol A (BPA) and dibutyl phthalate (DBP), on juxtacrine communication in adult rat testis, by evaluating the expression of Notch pathway components. Testicular explant were exposed in vitro to BPA (5 × 10-6 M, 2.5 × 10-5 M, 5 × 10-5 M) or DBP (10-6 M, 10-5 M, 10-4 M) for 24 h. To determine the expression of Notch1, Dll4, Hey1, Hes1 and Hey5 real-time RT-PCR was used. Protein levels and localization of NOTCH1 receptor, its ligand DLL4 as well as HEY1, HES1 and HEY5 factors were detected by western blot analysis and immunohistochemistry, respectively. Upregulation of Notch1, Dll4 and Hey1 at the mRNA and protein level was demonstrated in testis explants after BPA and DBP treatment (p < 0.05; p < 0.01; p < 0.001). Hes5 expression decreased after BPA (p < 0.05; p < 0.01; p < 0.001), whereas Hes1 expression was not altered by either BPA or DBP. Tested chemicals altered immunoexpression of activated NOTCH1, DLL4, HEY1 and HES5 both in seminiferous epithelium and interstitial tissue, exerting differential effects on particular cell types. In conclusion, BPA and DBP affect Notch signaling pathway in rat testis, which indicates that juxtacrine communication is a potential target for the action of plastic derived compounds in male gonad.
Collapse
Affiliation(s)
- Alicja Kamińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Laura Pardyak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Sylwia Marek
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Ewelina Górowska-Wójtowicz
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Kotula-Balak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Barbara Bilińska
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
33
|
Su DM, Feng Y, Wang L, Wu YL, Ge RS, Ma X. Influence of fetal Leydig cells on the development of adult Leydig cell population in rats. J Reprod Dev 2018. [PMID: 29515056 PMCID: PMC6021611 DOI: 10.1262/jrd.2017-102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Leydig cells are the main endogenous testosterone synthesis cells in the body. Testosterone is an essential hormone in males that affects metabolism, emotion, and pubertal development.
However, little is known about the development of Leydig cells and relationship between fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). The aims of this study were to investigate
the effect of (FLCs) on ALC development. Our study showed that FLCs in neonatal rat testis can be eliminated by 100 mg/kg ethane dimethane sulfonate (EDS) treatment without affecting the
health of newborn rats. Immunohistological results showed that eliminating FLCs led to early re-generation of the ALC population (progenitor Leydig cells [PLCs] and ALCs) accompanied at
first by increased and then by decreased serum testosterone, indicating that ALCs which appeared after neonatal EDS treatment were degenerated or had attenuated functions. Our results showed
that FLCs were eliminated 4 days after EDS treatment, the ALC population regenerated by 21 days, and serum testosterone levels dramatically decreased at 56 days. Collectively, our results
indicate that the ablation of FLCs in neonatal rat results in abnormal development of ALCs. Our study further indicates that abnormal development of Leydig cells in the fetal stage leads to
steroid hormone disorders, such as testosterone deficiency, in the adult stage. Therefore, studies of Leydig cell development are important for understanding the pathogenesis of testosterone
deficiency or pubertas praecox.
Collapse
Affiliation(s)
- Dong-Mei Su
- West China Hospital, West China School of Clinical Medicine, Sichuan University, Chengdu 610041, China
| | - Ying Feng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Lin Wang
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yi-Lun Wu
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ren-Shan Ge
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Xue Ma
- Department of Pediatric Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Svechnikov K, Savchuk I, Morvan ML, Antignac JP, Le Bizec B, Söder O. Phthalates Exert Multiple Effects on Leydig Cell Steroidogenesis. Horm Res Paediatr 2018; 86:253-263. [PMID: 26559938 DOI: 10.1159/000440619] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/21/2015] [Indexed: 11/19/2022] Open
Abstract
Humans are significantly exposed to phthalates via food packaging, cosmetics and medical devices such as tubings and catheters. Testicular Leydig cells (LCs) are suggested to be among the main targets of phthalate toxicity in the body. However, their sensitivity to phthalates is species-dependent. This paper describes the response of the LCs from different species (mouse, rat and human) to phthalate exposure in different experimental paradigms (in vivo, ex vivo and in vitro), with particular focus on mechanisms of phthalate action on LC steroidogenesis. A comprehensive analysis of the impact of phthalate diesters and phthalate monoesters on LCs in different stages of their development is presented and possible mechanisms of phthalates action are discussed. Finally novel, not yet fully elucidated sites of action of phthalate monoesters on the backdoor pathway of 5α-dihydrotestosterone biosynthesis in immature mouse LCs and their effects on steroidogenesis and redox state in adult mouse LCs are reported.
Collapse
Affiliation(s)
- Konstantin Svechnikov
- Department of Women's and Children's Health, Pediatric Endocrinology Unit, Karolinska Institute and University Hospital, Q2:08, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
35
|
MAFB is dispensable for the fetal testis morphogenesis and the maintenance of spermatogenesis in adult mice. PLoS One 2018; 13:e0190800. [PMID: 29324782 PMCID: PMC5764304 DOI: 10.1371/journal.pone.0190800] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 12/20/2017] [Indexed: 01/01/2023] Open
Abstract
The transcription factor MAFB is an important regulator of the development and differentiation of various organs and tissues. Previous studies have shown that MAFB is expressed in embryonic and adult mouse testes and is expected to act as the downstream target of retinoic acid (RA) to initiate spermatogenesis. However, its exact localization and function remain unclear. Here, we localized MAFB expression in embryonic and adult testes and analyzed its gene function using Mafb-deficient mice. We found that MAFB and c-MAF are the only large MAF transcription factors expressed in testes, while MAFA and NRL are not. MAFB was localized in Leydig and Sertoli cells at embryonic day (E) 18.5 but in Leydig cells, Sertoli cells, and pachytene spermatocytes in adults. Mafb-deficient testes at E18.5 showed fully formed seminiferous tubules with no abnormal structure or differences in testicular somatic cell numbers compared with those of control wild-type mice. Additionally, the expression levels of genes related to development and function of testicular cells were unchanged between genotypes. In adults, the expression of MAFB in Sertoli cells was shown to be stage specific and induced by RA. By generating Mafbfl/fl CAG-CreER™ (Mafb-cKO) mice, in which Cre recombinase was activated upon tamoxifen treatment, we found that the neonatal cKO mice died shortly upon Mafb deletion, but adult cKO mice were alive upon deletion. Adult cKO mice were fertile, and spermatogenesis maintenance was normal, as indicated by histological analysis, hormone levels, and germ cell stage-specific markers. Moreover, there were no differences in the proportion of seminiferous stages between cKO mice and controls. However, RNA-Seq analysis of cKO Sertoli cells revealed that the down-regulated genes were related to immune function and phagocytosis activity but not spermatogenesis. In conclusion, we found that MAFB is dispensable for fetal testis morphogenesis and spermatogenesis maintenance in adult mice, despite the significant gene expression in different cell types, but MAFB might be critical for phagocytosis activity of Sertoli cells.
Collapse
|
36
|
Skaftnesmo KO, Edvardsen RB, Furmanek T, Crespo D, Andersson E, Kleppe L, Taranger GL, Bogerd J, Schulz RW, Wargelius A. Integrative testis transcriptome analysis reveals differentially expressed miRNAs and their mRNA targets during early puberty in Atlantic salmon. BMC Genomics 2017; 18:801. [PMID: 29047327 PMCID: PMC5648517 DOI: 10.1186/s12864-017-4205-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Our understanding of the molecular mechanisms implementing pubertal maturation of the testis in vertebrates is incomplete. This topic is relevant in Atlantic salmon aquaculture, since precocious male puberty negatively impacts animal welfare and growth. We hypothesize that certain miRNAs modulate mRNAs relevant for the initiation of puberty. To explore which miRNAs regulate mRNAs during initiation of puberty in salmon, we performed an integrated transcriptome analysis (miRNA and mRNA-seq) of salmon testis at three stages of development: an immature, long-term quiescent stage, a prepubertal stage just before, and a pubertal stage just after the onset of single cell proliferation activity in the testis. RESULTS Differentially expressed miRNAs clustered into 5 distinct expression profiles related to the immature, prepubertal and pubertal salmon testis. Potential mRNA targets of these miRNAs were predicted with miRmap and filtered for mRNAs displaying negatively correlated expression patterns. In summary, this analysis revealed miRNAs previously known to be regulated in immature vertebrate testis (miR-101, miR-137, miR-92b, miR-18a, miR-20a), but also miRNAs first reported here as regulated in the testis (miR-new289, miR-30c, miR-724, miR-26b, miR-new271, miR-217, miR-216a, miR-135a, miR-new194 and the novel predicted n268). By KEGG enrichment analysis, progesterone signaling and cell cycle pathway genes were found regulated by these differentially expressed miRNAs. During the transition into puberty we found differential expression of miRNAs previously associated (let7a/b/c), or newly associated (miR-15c, miR-2184, miR-145 and the novel predicted n7a and b) with this stage. KEGG enrichment analysis revealed that mRNAs of the Wnt, Hedgehog and Apelin signaling pathways were potential regulated targets during the transition into puberty. Likewise, several regulated miRNAs in the pubertal stage had earlier been associated (miR-20a, miR-25, miR-181a, miR-202, let7c/d/a, miR-125b, miR-222a/b, miR-190a) or have now been found connected (miR-2188, miR-144, miR-731, miR-8157 and the novel n2) to the initiation of puberty. CONCLUSIONS This study has - for the first time - linked testis maturation to specific miRNAs and their inversely correlated expressed targets in Atlantic salmon. The study indicates a broad functional conservation of already known miRNAs and associated pathways involved in the transition into puberty in vertebrates. The analysis also reveals miRNAs not previously associated with testis tissue or its maturation, which calls for further functional studies in the testis.
Collapse
Affiliation(s)
- K O Skaftnesmo
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway.
| | - R B Edvardsen
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - T Furmanek
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - D Crespo
- Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - E Andersson
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - L Kleppe
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - G L Taranger
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| | - J Bogerd
- Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - R W Schulz
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway.,Reproductive Biology group, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - A Wargelius
- Institute of Marine Research, Postboks 1870 Nordnes, 5817, Bergen, Norway
| |
Collapse
|
37
|
Ye L, Li X, Li L, Chen H, Ge RS. Insights into the Development of the Adult Leydig Cell Lineage from Stem Leydig Cells. Front Physiol 2017; 8:430. [PMID: 28701961 PMCID: PMC5487449 DOI: 10.3389/fphys.2017.00430] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/06/2017] [Indexed: 02/06/2023] Open
Abstract
Adult Leydig cells (ALCs) are the steroidogenic cells in the testes that produce testosterone. ALCs develop postnatally from a pool of stem cells, referred to as stem Leydig cells (SLCs). SLCs are spindle-shaped cells that lack steroidogenic cell markers, including luteinizing hormone (LH) receptor and 3β-hydroxysteroid dehydrogenase. The commitment of SLCs into the progenitor Leydig cells (PLCs), the first stage in the lineage, requires growth factors, including Dessert Hedgehog (DHH) and platelet-derived growth factor-AA. PLCs are still spindle-shaped, but become steroidogenic and produce mainly androsterone. The next transition in the lineage is from PLC to the immature Leydig cell (ILC). This transition requires LH, DHH, and androgen. ILCs are ovoid cells that are competent for producing a different form of androgen, androstanediol. The final stage in the developmental lineage is ALC. The transition to ALC involves the reduced expression of 5α-reductase 1, a step that is necessary to make the cells to produce testosterone as the final product. The transitions along the Leydig cell lineage are associated with the progressive down-regulation of the proliferative activity, and the up-regulation of steroidogenic capacity, with each step requiring unique regulatory signaling.
Collapse
Affiliation(s)
- Leping Ye
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Xiaoheng Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Haolin Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Ren-Shan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| |
Collapse
|
38
|
Sun X, Ito J, Potter SJ, Dey SK, DeFalco T. Extragonadal oocytes residing in the mouse ovarian hilum contribute to fertility. Biol Reprod 2017; 96:1060-1070. [PMID: 28339687 PMCID: PMC6279060 DOI: 10.1095/biolreprod.116.145631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/13/2017] [Accepted: 02/13/2017] [Indexed: 12/17/2022] Open
Abstract
The observation of pups born from recipient and donor mice after ovariectomy followed by ovarian transplant poses the interesting possibility of an extraovarian source of oocytes. However, whether mammalian adult oocytes reside in extragonadal tissues remains elusive. Using transgenic fluorescent reporter mice and transplantation surgeries, we demonstrate the presence of both donor- and recipient-derived corpora lutea and recovery of both donor- and recipient-derived offspring from ovariectomized mice after transplantation of donor ovaries. A potential region for extraovarian oocytes is the hilum, a ligament-like structure between the ovary and the reproductive tract. Immunofluorescent confocal microscopy of mouse ovaries and reproductive tracts revealed that a population of primordial follicles resides outside the ovary within the hilum. Ovariectomy-only controls confirmed that oocytes remain in the recipient hilum after surgery. These results provide evidence that the hilum is a reserve source of follicles, which likely return to the ovary for maturation and ovulation. By identifying a new follicle reservoir, our study addresses a long-standing question in reproductive biology and contributes to new conceptual knowledge about ovarian function and fertility.
Collapse
Affiliation(s)
- Xiaofei Sun
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Junya Ito
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sarah J. Potter
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sudhansu K. Dey
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
39
|
Shima Y, Morohashi KI. Leydig progenitor cells in fetal testis. Mol Cell Endocrinol 2017; 445:55-64. [PMID: 27940302 DOI: 10.1016/j.mce.2016.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/18/2016] [Accepted: 12/07/2016] [Indexed: 12/21/2022]
Abstract
Testicular Leydig cells play pivotal roles in masculinization of organisms by producing androgens. At least two distinct Leydig cell populations sequentially emerge in the mammalian testis. Leydig cells in the fetal testis (fetal Leydig cells) appear just after initial sex differentiation and induce masculinization of male fetuses. Although there has been a debate on the fate of fetal Leydig cells in the postnatal testis, it has been generally believed that fetal Leydig cells regress and are completely replaced by another Leydig cell population, adult Leydig cells. Recent studies revealed that gene expression patterns are different between fetal and adult Leydig cells and that the androgens produced in fetal Leydig cells are different from those in adult Leydig cells in mice. Although these results suggested that fetal and adult Leydig cells have distinct origins, several recent studies of mouse models support the hypothesis that fetal and adult Leydig cells arise from a common progenitor pool. In this review, we first provide an overview of previous knowledge, mainly from mouse studies, focusing on the cellular origins of fetal Leydig cells and the regulatory mechanisms underlying fetal Leydig cell differentiation. In addition, we will briefly discuss the functional differences of fetal Leydig cells between human and rodents. We will also discuss recent studies with mouse models that give clues for understanding how the progenitor cells in the fetal testis are subsequently destined to become fetal or adult Leydig cells.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of Anatomy, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | - Ken-Ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
40
|
Lin YT, Barske L, DeFalco T, Capel B. Numb regulates somatic cell lineage commitment during early gonadogenesis in mice. Development 2017; 144:1607-1618. [PMID: 28360133 DOI: 10.1242/dev.149203] [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: 01/16/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022]
Abstract
During early gonadogenesis, proliferating cells in the coelomic epithelium (CE) give rise to most of the somatic cells in both XX and XY gonads. Previous dye-labeling experiments showed that a single CE cell could give rise to additional CE cells and to both supporting and interstitial cell lineages, implying that cells in the CE domain are multipotent progenitors, and suggesting that an asymmetric division is involved in the acquisition of gonadal cell fates. We found that NUMB is asymmetrically localized in CE cells, suggesting that it might be involved. To test this hypothesis, we conditionally deleted Numb on a Numbl mutant background just prior to gonadogenesis. Mutant gonads showed a loss of cell polarity in the surface epithelial layers, large interior cell patches expressing the undifferentiated cell marker LHX9, and a loss of differentiated cells in somatic cell lineages. These results indicate that NUMB is necessary for establishing polarity in CE cells, and that asymmetric divisions resulting from CE polarity are required for commitment to differentiated somatic cell fates. Surprisingly, germ cells, which do not arise from the CE, were also affected in mutants, which may be a direct or indirect effect of loss of Numb.
Collapse
Affiliation(s)
- Yi-Tzu Lin
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lindsey Barske
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Blanche Capel
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
41
|
Wen Q, Wang Y, Tang J, Cheng CY, Liu YX. Sertoli Cell Wt1 Regulates Peritubular Myoid Cell and Fetal Leydig Cell Differentiation during Fetal Testis Development. PLoS One 2016; 11:e0167920. [PMID: 28036337 PMCID: PMC5201236 DOI: 10.1371/journal.pone.0167920] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
Sertoli cells play a significant role in regulating fetal testis compartmentalization to generate testis cords and interstitium during development. The Sertoli cell Wilms’ tumor 1 (Wt1) gene, which encodes ~24 zinc finger-containing transcription factors, is known to play a crucial role in fetal testis cord assembly and maintenance. However, whether Wt1 regulates fetal testis compartmentalization by modulating the development of peritubular myoid cells (PMCs) and/or fetal Leydig cells (FLCs) remains unknown. Using a Wt1-/flox; Amh-Cre mouse model by deleting Wt1 in Sertoli cells (Wt1SC-cKO) at embryonic day 14.5 (E14.5), Wt1 was found to regulate PMC and FLC development. Wt1 deletion in fetal testis Sertoli cells caused aberrant differentiation and proliferation of PMCs, FLCs and interstitial progenitor cells from embryo to newborn, leading to abnormal fetal testis interstitial development. Specifically, the expression of PMC marker genes α-Sma, Myh11 and Des, and interstitial progenitor cell marker gene Vcam1 were down-regulated, whereas FLC marker genes StAR, Cyp11a1, Cyp17a1 and Hsd3b1 were up-regulated, in neonatal Wt1SC-cKO testes. The ratio of PMC:FLC were also reduced in Wt1SC-cKO testes, concomitant with a down-regulation of Notch signaling molecules Jag 1, Notch 2, Notch 3, and Hes1 in neonatal Wt1SC-cKO testes, illustrating changes in the differentiation status of FLC from their interstitial progenitor cells during fetal testis development. In summary, Wt1 regulates the development of FLC and interstitial progenitor cell lineages through Notch signaling, and it also plays a role in PMC development. Collectively, these effects confer fetal testis compartmentalization.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
| | - Yuqian Wang
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jixin Tang
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - C. Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, United States of America
- * E-mail: (YXL); (CYC)
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- * E-mail: (YXL); (CYC)
| |
Collapse
|
42
|
Crespo D, Assis LHC, Furmanek T, Bogerd J, Schulz RW. Expression profiling identifies Sertoli and Leydig cell genes as Fsh targets in adult zebrafish testis. Mol Cell Endocrinol 2016; 437:237-251. [PMID: 27566230 DOI: 10.1016/j.mce.2016.08.033] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/27/2016] [Accepted: 08/22/2016] [Indexed: 11/26/2022]
Abstract
Spermatogonial stem cells are quiescent, undergo self-renewal or differentiating divisions, thereby forming the cellular basis of spermatogenesis. This cellular development is orchestrated by follicle-stimulating hormone (FSH), through the production of Sertoli cell-derived factors, and by Leydig cell-released androgens. Here, we investigate the transcriptional events induced by Fsh in a steroid-independent manner on the restart of zebrafish (Danio rerio) spermatogenesis ex vivo, using testis from adult males where type A spermatogonia were enriched by estrogen treatment in vivo. Under these conditions, RNA sequencing preferentially detected differentially expressed genes in somatic/Sertoli cells. Fsh-stimulated spermatogonial proliferation was accompanied by modulating several signaling systems (i.e. Tgf-β, Hedgehog, Wnt and Notch pathways). In silico protein-protein interaction analysis indicated a role for Hedgehog family members potentially integrating signals from different pathways during fish spermatogenesis. Moreover, Fsh had a marked impact on metabolic genes, such as lactate and fatty acid metabolism, or on Sertoli cell barrier components. Fish Leydig cells express the Fsh receptor and one of the most robust Fsh-responsive genes was insulin-like 3 (insl3), a Leydig cell-derived growth factor. Follow-up work showed that recombinant zebrafish Insl3 mediated pro-differentiation effects of Fsh on spermatogonia in an androgen-independent manner. Our experimental approach allowed focusing on testicular somatic genes in zebrafish and showed that the activity of signaling systems known to be relevant in stem cells was modulated by Fsh, providing promising leads for future work, as exemplified by the studies on Insl3.
Collapse
Affiliation(s)
- Diego Crespo
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Luiz H C Assis
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Tomasz Furmanek
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands; Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway.
| |
Collapse
|
43
|
Chen H, Jin S, Huang S, Folmer J, Liu J, Ge R, Zirkin BR. Transplantation of alginate-encapsulated seminiferous tubules and interstitial tissue into adult rats: Leydig stem cell differentiation in vivo? Mol Cell Endocrinol 2016; 436:250-8. [PMID: 27591121 PMCID: PMC5050555 DOI: 10.1016/j.mce.2016.08.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/09/2016] [Accepted: 08/29/2016] [Indexed: 01/06/2023]
Abstract
In vivo and in vitro studies were conducted to determine whether testosterone-producing Leydig cells are able to develop from cells associated with rat seminiferous tubules, interstitium, or both. Adult rat seminiferous tubules and interstitium were isolated, encapsulated separately in alginate, and implanted subcutaneously into castrated rats. With implanted tubules, serum testosterone increased through two months. Tubules removed from the implanted rats and incubated with LH produced testosterone, and cells on the tubule surfaces expressed steroidogenic enzymes. With implanted interstitial tissue, serum levels of testosterone remained undetectable. However, co-culture of interstitium plus tubules in vitro resulted in the formation of Leydig cells by both compartments. These results indicate that seminiferous tubules contain both cellular and paracrine factors necessary for the differentiation of Leydig cells, and that the interstitial compartment contains precursor cells capable of forming testosterone-producing Leydig cells but requires stimulation by paracrine factors from the seminiferous tubules to do so.
Collapse
Affiliation(s)
- Haolin Chen
- Center for Scientific Research, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shiying Jin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shengsong Huang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Urology, Tongji Hospital, Tongji University School of Medicine, Putuo, Shanghai 200065, China
| | - Janet Folmer
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - June Liu
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Renshan Ge
- Center for Scientific Research, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Barry R Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
44
|
Wen Q, Cheng CY, Liu YX. Development, function and fate of fetal Leydig cells. Semin Cell Dev Biol 2016; 59:89-98. [PMID: 26968934 PMCID: PMC5016207 DOI: 10.1016/j.semcdb.2016.03.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/21/2022]
Abstract
During fetal testis development, fetal Leydig cells (FLCs) are found to be originated from multiple progenitor cells. FLC specification and function are under tight regulation of specific genes and signaling proteins. Furthermore, Sertoli cells play a crucial role to regulate FLC differentiation during fetal testis development. FLC progenitor- and FLC-produced biomolecules are also involved in the differentiation and activity of rodent FLCs. The main function of FLCs is to produce androgens to masculinize XY embryos. However, FLCs are capable of producing androstenedione but not testosterone due to the lack of 17β-HSD (17β-hydroxysteroid dehydrogenase), but fetal Sertoli cells express 17β-HSD which thus transforms androstenedione to testosterone in the fetal testis. On the other hand, FLCs produce activin A to regulate Sertoli cell proliferation, and Sertoli cells in turn modulate testis cord expansion. It is now generally accepted that adult Leydig cells (ALCs) gradually replace FLCs during postnatal development to produce testosterone to support spermatogenesis as FLCs undergo degeneration in neonatal and pre-pubertal testes. However, based on studies using genetic tracing mouse models, FLCs are found to persist in adult testes, making up ∼20% of total Leydig cells. In this review, we evaluate the latest findings regarding the development, function and fate of FLCs during fetal and adult testis development.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York 10065, United States.
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
45
|
Anesetti G, Chávez-Genaro R. Ovarian follicular dynamics after aromatizable or non aromatizable neonatal androgenization. J Mol Histol 2016; 47:491-501. [PMID: 27541036 DOI: 10.1007/s10735-016-9692-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/10/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Gabriel Anesetti
- Histology and Embryology Department, School of Medicine, General Flores 2125, CP 11800, Montevideo, Uruguay.
| | - Rebeca Chávez-Genaro
- Histology and Embryology Department, School of Medicine, General Flores 2125, CP 11800, Montevideo, Uruguay
| |
Collapse
|
46
|
Campbell JL, Andersen ME, Hinderliter PM, Yi KD, Pastoor TP, Breckenridge CB, Clewell HJ. PBPK Model for Atrazine and Its Chlorotriazine Metabolites in Rat and Human. Toxicol Sci 2016; 150:441-53. [PMID: 26794140 PMCID: PMC4809456 DOI: 10.1093/toxsci/kfw014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The previously-published physiologically based pharmacokinetic model for atrazine (ATZ), deisopropylatrazine (DIA), deethylatrazine (DEA), and diaminochlorotriazine (DACT), which collectively comprise the total chlorotriazines (TCT) as represented in this study, was modified to allow for scaling to humans. Changes included replacing the fixed dose-dependent oral uptake rates with a method that represented delayed absorption observed in rats administered ATZ as a bolus dose suspended in a methylcellulose vehicle. Rate constants for metabolism of ATZ to DIA and DEA, followed by metabolism of DIA and DEA to DACT were predicted using a compartmental model describing the metabolism of the chlorotriazines by rat and human hepatocytesin vitro Overall, the model successfully predicted both the 4-day plasma time-course data in rats administered ATZ by bolus dose (3, 10, and 50 mg/kg/day) or in the diet (30, 100, or 500 ppm). Simulated continuous daily exposure of a 55-kg adult female to ATZ at a dose of 1.0 µg/kg/day resulted in steady-state urinary concentrations of 0.6, 1.4, 2.5, and 6.0 µg/L for DEA, DIA, DACT, and TCT, respectively. The TCT (ATZ + DEA + DIA + DACT) human urinary biomonitoring equivalent concentration following continuous exposure to ATZ at the chronic point of departure (POD = 1.8 mg/kg/day) was 360.6 μg/L.
Collapse
Affiliation(s)
- Jerry L Campbell
- *The Hamner Institutes for Health Sciences, Center for Human Health Assessment, Research Triangle Park, North Carolina 27709-2137;
| | - Melvin E Andersen
- *The Hamner Institutes for Health Sciences, Center for Human Health Assessment, Research Triangle Park, North Carolina 27709-2137
| | | | - Kun Don Yi
- Syngenta Crop Protection, LLC, Greensboro, North Carolina 27419-8300
| | - Timothy P Pastoor
- Pastoor Science Communications, LLC, Greensboro, North Carolina 27455-3415
| | | | - Harvey J Clewell
- *The Hamner Institutes for Health Sciences, Center for Human Health Assessment, Research Triangle Park, North Carolina 27709-2137
| |
Collapse
|
47
|
Potter SJ, Kumar DL, DeFalco T. Origin and Differentiation of Androgen-Producing Cells in the Gonads. Results Probl Cell Differ 2016; 58:101-134. [PMID: 27300177 DOI: 10.1007/978-3-319-31973-5_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Sexual reproduction is dependent on the activity of androgenic steroid hormones to promote gonadal development and gametogenesis. Leydig cells of the testis and theca cells of the ovary are critical cell types in the gonadal interstitium that carry out steroidogenesis and provide key androgens for reproductive organ function. In this chapter, we will discuss important aspects of interstitial androgenic cell development in the gonad, including: the potential cellular origins of interstitial steroidogenic cells and their progenitors; the molecular mechanisms involved in Leydig cell specification and differentiation (including Sertoli-cell-derived signaling pathways and Leydig-cell-related transcription factors and nuclear receptors); the interactions of Leydig cells with other cell types in the adult testis, such as Sertoli cells, germ cells, peritubular myoid cells, macrophages, and vascular endothelial cells; the process of steroidogenesis and its systemic regulation; and a brief discussion of the development of theca cells in the ovary relative to Leydig cells in the testis. Finally, we will describe the dynamics of steroidogenic cells in seasonal breeders and highlight unique aspects of steroidogenesis in diverse vertebrate species. Understanding the cellular origins of interstitial steroidogenic cells and the pathways directing their specification and differentiation has implications for the study of multiple aspects of development and will help us gain insights into the etiology of reproductive system birth defects and infertility.
Collapse
Affiliation(s)
- Sarah J Potter
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Deepti Lava Kumar
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
48
|
Karpova T, Ravichandiran K, Insisienmay L, Rice D, Agbor V, Heckert LL. Steroidogenic factor 1 differentially regulates fetal and adult leydig cell development in male mice. Biol Reprod 2015; 93:83. [PMID: 26269506 DOI: 10.1095/biolreprod.115.131193] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022] Open
Abstract
The nuclear receptor steroidogenic factor 1 (SF-1, AD4BP, NR5A1) is a key regulator of the endocrine axes and is essential for adrenal and gonad development. Partial rescue of Nr5a1(-/-) mice with an SF-1-expressing transgene caused a hypomorphic phenotype that revealed its roles in Leydig cell development. In contrast to controls, all male rescue mice (Nr5a1(-/-);tg(+/0)) showed varying signs of androgen deficiency, including spermatogenic arrest, cryptorchidism, and poor virilization. Expression of various Leydig cell markers measured by immunohistochemistry, Western blot analysis, and RT-PCR indicated fetal and adult Leydig cell development were differentially impaired. Whereas fetal Leydig cell development was delayed in Nr5a1(-/-);tg(+/0) embryos, it recovered to control levels by birth. In contrast, Sult1e1, Vcam1, and Hsd3b6 transcript levels in adult rescue testes indicated complete blockage in adult Leydig cell development. In addition, between Postnatal Days 8 and 12, peritubular cells expressing PTCH1, SF-1, and CYP11A1 were observed in control testes but not in rescue testes, indicating SF-1 is needed for either survival or differentiation of adult Leydig cell progenitors. Cultured prepubertal rat peritubular cells also expressed SF-1 and PTCH1, but Cyp11a1 was expressed only after treatment with cAMP and retinoic acid. Together, data show SF-1 is needed for proper development of fetal and adult Leydig cells but with distinct primary functions; in fetal Leydig cells, it regulates differentiation, whereas in adult Leydig cells it regulates progenitor cell formation and/or survival.
Collapse
Affiliation(s)
- Tatiana Karpova
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Kumarasamy Ravichandiran
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lovella Insisienmay
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Daren Rice
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Valentine Agbor
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Leslie L Heckert
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
49
|
Macrophages Contribute to the Spermatogonial Niche in the Adult Testis. Cell Rep 2015; 12:1107-19. [PMID: 26257171 DOI: 10.1016/j.celrep.2015.07.015] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/02/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023] Open
Abstract
The testis produces sperm throughout the male reproductive lifespan by balancing self-renewal and differentiation of spermatogonial stem cells (SSCs). Part of the SSC niche is thought to lie outside the seminiferous tubules of the testis; however, specific interstitial components of the niche that regulate spermatogonial divisions and differentiation remain undefined. We identified distinct populations of testicular macrophages, one of which lies on the surface of seminiferous tubules, in close apposition to areas of tubules enriched for undifferentiated spermatogonia. These macrophages express spermatogonial proliferation- and differentiation-inducing factors, such as colony-stimulating factor 1 (CSF1) and enzymes involved in retinoic acid (RA) biosynthesis. We show that transient depletion of macrophages leads to a disruption in spermatogonial differentiation. These findings reveal an unexpected role for macrophages in the spermatogonial niche in the testis and raise the possibility that macrophages play previously unappreciated roles in stem/progenitor cell regulation in other tissues.
Collapse
|
50
|
George RM, Hahn KL, Rawls A, Viger RS, Wilson-Rawls J. Notch signaling represses GATA4-induced expression of genes involved in steroid biosynthesis. Reproduction 2015; 150:383-94. [PMID: 26183893 DOI: 10.1530/rep-15-0226] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022]
Abstract
Notch2 and Notch3 and genes of the Notch signaling network are dynamically expressed in developing follicles, where they are essential for granulosa cell proliferation and meiotic maturation. Notch receptors, ligands, and downstream effector genes are also expressed in testicular Leydig cells, predicting a potential role in regulating steroidogenesis. In this study, we sought to determine if Notch signaling in small follicles regulates the proliferation response of granulosa cells to FSH and represses the up-regulation steroidogenic gene expression that occurs in response to FSH as the follicle grows. Inhibition of Notch signaling in small preantral follicles led to the up-regulation of the expression of genes in the steroid biosynthetic pathway. Similarly, progesterone secretion by MA-10 Leydig cells was significantly inhibited by constitutively active Notch. Together, these data indicated that Notch signaling inhibits steroidogenesis. GATA4 has been shown to be a positive regulator of steroidogenic genes, including STAR protein, P450 aromatase, and 3B-hydroxysteroid dehydrogenase. We observed that Notch downstream effectors HEY1, HEY2, and HEYL are able to differentially regulate these GATA4-dependent promoters. These data are supported by the presence of HEY/HES binding sites in these promoters. These studies indicate that Notch signaling has a role in the complex regulation of the steroidogenic pathway.
Collapse
Affiliation(s)
- Rajani M George
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Katherine L Hahn
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Alan Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Robert S Viger
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4 School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Jeanne Wilson-Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| |
Collapse
|