Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Commun Biol ; 5(1): 974, 2022 09 15.
Article En | MEDLINE | ID: mdl-36109592

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 ß10 (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.


Androgens , MAP Kinase Signaling System , Androgens/metabolism , Fetus , Humans , Male , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thymosin , ras Proteins/metabolism
2.
Front Cell Dev Biol ; 10: 907989, 2022.
Article En | MEDLINE | ID: mdl-35573691

Primordial germ cells nesting in the fetal testis give rise to gonocytes. The gonocytes then transform into spermatogenic stem cells (SSCs) during the neonatal period and thereafter serve as a lifetime source of spermatogenesis. Therefore, gonocyte to SSC transformation is quite an important process that supports fertility in males. During the gonocyte to SSC transformation, morphological and transcriptomic changes sequentially occur and gonocytes migrate from the center to the peripheral region of the seminiferous tubules. However, extrinsic signals which trigger the transcriptomic changes as well as the migration are not yet fully clarified. Recent studies have drawn attention to the temporal activation of the hypothalamic-pituitary-gonadal axis during the neonatal stage which occurs concurrently with SSC formation. This phenomenon is called mini-puberty, and recent studies on human cryptorchid patients as well as animal models partially support the hypothesis that mini-puberty plays pivotal roles in gonocyte-to-SSC transformation. Focusing on this point, here, we aimed to discuss the latest knowledge on the importance of mini-puberty in spermatogenesis in this review.

3.
Microscopy (Oxf) ; 71(3): 187-192, 2022 Jun 06.
Article En | MEDLINE | ID: mdl-35325180

Focused ion beamed (FIB) SEM has a higher spatial resolution than other volume-imaging methods owing to the use of ion beams. However, in this method, it is challenging to analyse entire biological structures buried deep in the resin block. We developed a novel volume-imaging method by combining array tomography and FIB-SEM tomography and investigated the chondrocyte ultrastructure. Our method imparts certainty in determining the analysis area such that cracks or areas with poor staining within the block are avoided. The chondrocyte surface showed fine dendritic processes that were thinner than ultrathin sections. Upon combination with immunostaining, this method holds promise for analysing mesoscopic architectures.


Deceleration , Tomography , Imaging, Three-Dimensional/methods , Microscopy, Electron, Scanning , Tomography, X-Ray Computed
4.
Int J Mol Sci ; 24(1)2022 Dec 22.
Article En | MEDLINE | ID: mdl-36613635

Nuclear receptor subfamily 5 group A member 1 (NR5A1) is expressed in the pituitary gonadotrope and regulates their differentiation. Although several regulatory regions were implicated in Nr5a1 gene expression in the pituitary gland, none of these regions have been verified using mouse models. Furthermore, the molecular functions of NR5A1 in the pituitary gonadotrope have not been fully elucidated. In the present study, we generated mice lacking the pituitary enhancer located in the 6th intron of the Nr5a1 gene. These mice showed pituitary gland-specific disappearance of NR5A1, confirming the functional importance of the enhancer. Enhancer-deleted male mice demonstrated no defects at fetal stages. Meanwhile, androgen production decreased markedly in adult, and postnatal development of reproductive organs, such as the seminal vesicle, prostate, and penis was severely impaired. We further performed transcriptomic analyses of the whole pituitary gland of the enhancer-deleted mice and controls, as well as gonadotropes isolated from Ad4BP-BAC-EGFP mice. These analyses identified several genes showing gonadotrope-specific, NR5A1-dependent expressions, such as Spp1, Tgfbr3l, Grem1, and Nr0b2. These factors are thought to function downstream of NR5A1 and play important roles in reproductive organ development through regulation of pituitary gonadotrope functions.


Gonadotrophs , Pituitary Gland , Regulatory Sequences, Nucleic Acid , Steroidogenic Factor 1 , Animals , Male , Mice , Gonadotrophs/metabolism , Introns/genetics , Pituitary Gland/metabolism , Steroidogenic Factor 1/genetics
5.
Science ; 373(6552)2021 07 16.
Article En | MEDLINE | ID: mdl-34437124

Oocytes mature in a specialized fluid-filled sac, the ovarian follicle, which provides signals needed for meiosis and germ cell growth. Methods have been developed to generate functional oocytes from pluripotent stem cell-derived primordial germ cell-like cells (PGCLCs) when placed in culture with embryonic ovarian somatic cells. In this study, we developed culture conditions to recreate the stepwise differentiation process from pluripotent cells to fetal ovarian somatic cell-like cells (FOSLCs). When FOSLCs were aggregated with PGCLCs derived from mouse embryonic stem cells, the PGCLCs entered meiosis to generate functional oocytes capable of fertilization and development to live offspring. Generating functional mouse oocytes in a reconstituted ovarian environment provides a method for in vitro oocyte production and follicle generation for a better understanding of mammalian reproduction.


Mouse Embryonic Stem Cells/physiology , Oocytes/physiology , Oogenesis , Ovarian Follicle/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Embryonic Development , Female , Fertilization in Vitro , Male , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Inbred ICR , Mouse Embryonic Stem Cells/cytology , Oocytes/cytology , Ovarian Follicle/embryology , Ovarian Follicle/physiology , RNA-Seq , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Transcriptome
6.
Dev Psychobiol ; 63(4): 725-733, 2021 05.
Article En | MEDLINE | ID: mdl-33070342

Testosterone masculinizes male sexual behavior by providing organizational and activational effects during the perinatal and peripubertal periods and during adulthood, respectively. We revealed that the emission of ultrasonic vocalizations (USVs) and mounting behavior was regulated by different neural circuits. However, the detailed testosterone effects on these two behaviors have not been fully elucidated. Here, we evaluated the time-dependent effects of testosterone on USVs and mounting behavior in mice using a testosterone treatment model, in which females were treated with testosterone to assess the "gain-of-function" and a "loss-of-function" model. In the loss-of-function model, we used Ad4BP/SF-1ΔFLC/- male mice, in which testosterone production was abolished in prenatal and postnatal stages, and Ad4BP/SF-1ΔFLC/ΔFLC mice, in which testosterone production was markedly reduced only in prenatal stages. When testosterone was administered to female mice during the neonatal and peripubertal periods, but not during adulthood, USV emissions increased. Conversely, testosterone treatment in adult female mice increased the mounting behavior, but not USVs. In Ad4BP/SF-1ΔFLC/- mice, USVs and mounting behavior was completely absent. Ad4BP/SF-1ΔFLC/ΔFLC male mice displayed equivalent levels of USVs but less mounting behavior. Collectively, these results suggest that testosterone has dual regulatory roles in USV emissions and mounting behavior.


Ultrasonics , Vocalization, Animal , Animals , Female , Male , Mice , Pregnancy , Testosterone/pharmacology , Vocalization, Animal/physiology
7.
Reprod Med Biol ; 18(4): 323-330, 2019 Oct.
Article En | MEDLINE | ID: mdl-31607792

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.

8.
Development ; 145(23)2018 12 05.
Article En | MEDLINE | ID: mdl-30518625

Previous studies have established that fetal Leydig cells (FLCs) and adult Leydig cells (ALCs) show distinct functional characteristics. However, the lineage relationship between FLCs and ALCs has not been clarified yet. Here, we reveal that a subset of FLCs dedifferentiate at fetal stages to give rise to ALCs at the pubertal stage. Moreover, the dedifferentiated cells contribute to the peritubular myoid cell and vascular pericyte populations in the neonatal testis, and these non-steroidogenic cells serve as potential ALC stem cells. We generated FLC lineage-specific Nr5a1 (Ad4BP/SF-1) gene-disrupted mice and mice lacking the fetal Leydig enhancer (FLE) of the Nr5a1 gene. Phenotypes of these mice support the conclusion that most of the ALCs arise from dedifferentiated FLCs, and that the FLE of the Nr5a1 gene is essential for both initial FLC differentiation and pubertal ALC redifferentiation.


Adult Stem Cells/cytology , Cell Dedifferentiation , Fetus/cytology , Leydig Cells/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Lineage , Enhancer Elements, Genetic/genetics , Fibrosis , Integrases/metabolism , Leydig Cells/metabolism , Male , Mice , Models, Biological , Phenotype , Sequence Deletion/genetics , Steroidogenic Factor 1/metabolism , Testis/cytology , Testis/transplantation
9.
Commun Biol ; 1: 18, 2018.
Article En | MEDLINE | ID: mdl-30271905

Housekeeping metabolic pathways such as glycolysis are active in all cell types. In addition, many types of cells are equipped with cell-specific metabolic pathways. To properly perform their functions, housekeeping and cell-specific metabolic pathways must function cooperatively. However, the regulatory mechanisms that couple metabolic pathways remain largely unknown. Recently, we showed that the steroidogenic cell-specific nuclear receptor Ad4BP/SF-1, which regulates steroidogenic genes, also regulates housekeeping glycolytic genes. Here, we identify cholesterogenic genes as the targets of Ad4BP/SF-1. Further, we reveal that Ad4BP/SF-1 regulates Hummr, a candidate mediator of cholesterol transport from endoplasmic reticula to mitochondria. Given that cholesterol is the starting material for steroidogenesis and is synthesized from acetyl-CoA, which partly originates from glucose, our results suggest that multiple biological processes involved in synthesizing steroid hormones are governed by Ad4BP/SF-1. To our knowledge, this study provides the first example where housekeeping and cell-specific metabolism are coordinated at the transcriptional level.

10.
Genes Cells ; 23(11): 952-962, 2018 Nov.
Article En | MEDLINE | ID: mdl-30178547

FEAT, the protein encoded by methyltransferase-like 13 (METTL13), is aberrantly upregulated in most human cancers and potently drives tumorigenesis in vivo; however, its role in normal tissues remains elusive. Immunoblotting has displayed weak FEAT expression in normal human tissues, including the testis. Here, we found that FEAT is expressed in fetal and adult Leydig cells in the testis. FEAT knockdown using siRNA increased primary cilia formation in MA-10 Leydig tumor cells, accompanied by enhanced 5' adenosine monophosphate-activated protein kinase (AMPK) activation. Immunofluorescence analyses of FEAT-silenced MA-10 cells showed diminished insulin-like factor 3 (INSL3) expression. A male Mettl13+/- mouse developed bilateral intraabdominal cryptorchidism, suggesting defective INSL3 production by fetal Leydig cells. Leydig cells from the mouse showed markedly decreased INSL3 protein by immunohistochemistry. Together, these results suggest that FEAT facilitates the INSL3 production in testicular Leydig cells that is essential for transabdominal testis migration.


Cryptorchidism/metabolism , Insulin/metabolism , Leydig Cells/metabolism , Methyltransferases/metabolism , Proteins/metabolism , Testis/metabolism , Animals , Cell Movement , Cryptorchidism/pathology , Insulin/genetics , Leydig Cells/cytology , Male , Mice , Proteins/genetics , Testis/cytology , Transcriptional Activation
12.
Sci Rep ; 7: 41912, 2017 02 02.
Article En | MEDLINE | ID: mdl-28150810

SRY, a sex-determining gene, induces testis development in chromosomally female (XX) individuals. However, mouse XX Sertoli cells carrying Sry (XX/Sry Sertoli cells) are incapable of fully supporting germ cell development, even when the karyotype of the germ cells is XY. While it has therefore been assumed that XX/Sry Sertoli cells are not functionally equivalent to XY Sertoli cells, it has remained unclear which specific functions are affected. To elucidate the functional difference, we compared the gene expression of XY and XX/Sry Sertoli cells. Lactate and cholesterol metabolisms, essential for nursing the developing germ cells, were down-regulated in XX/Sry cells, which appears to be caused at least in part by the differential expression of histone modification enzymes SMCX/SMCY (H3K4me3 demethylase) and UTX/UTY (H3K27me3 demethylase) encoded by the sex chromosomes. We suggest that down-regulation of lactate and cholesterol metabolism that may be due to altered epigenetic modification affects the nursing functions of XX/Sry Sertoli cells.


Cholesterol/metabolism , Disorders of Sex Development/metabolism , Karyotype , Lactic Acid/metabolism , Sertoli Cells/metabolism , Animals , Cells, Cultured , Disorders of Sex Development/genetics , Female , Histone Code , Histone Demethylases/genetics , Histone Demethylases/metabolism , Male , Mice , Sertoli Cells/pathology , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , X Chromosome/genetics , Y Chromosome/genetics
13.
Endocr J ; 64(3): 315-324, 2017 Mar 31.
Article En | MEDLINE | ID: mdl-28202838

Ad4-binding protein/steroidogenic factor 1 (Ad4BP/SF-1), a member of the nuclear receptor superfamily, is expressed in steroidogenic cells and regulates all steroidogenic gene expression. We recently employed mRNA and chromatin immunoprecipitation sequence (ChIP-seq) to demonstrate that Ad4BP/SF-1 directly regulates the expression of nearly all glycolytic genes. The pentose phosphate pathway (PPP) contributes to the production of nicotinamide adenine dinucleotide phosphate (NADPH). Although the expression of PPP genes and intracellular NADPH were decreased by Ad4BP/SF-1 knockdown, these genes were not the direct targets of Ad4BP/SF-1. This study therefore investigates whether Ad4BP/SF-1 directly regulates genes implicated in NADPH production. Examination of previously published data sets of mRNA sequence (mRNA-seq) and ChIP-seq strongly suggested a possibility that other NADPH-producing genes, such as malic enzyme 1 (Me1) and methylenetetrahydrofolate dehydrogenase 2 (Mthfd2), are the direct targets of Ad4BP/SF-1. Reporter gene assays and determination of intracellular NADPH concentration supported the notion that Ad4BP/SF-1 regulates NADPH production by regulating these genes. NADPH is required for macromolecule synthesis of compounds such as steroids, and for detoxification of reactive oxygen species. When synthesizing steroid hormones, steroidogenic cells consume NADPH through enzymatic reactions mediated by steroidogenic P450s. NADPH is also consumed through elimination of reactive oxygen species produced as the byproducts of the P450 reactions. Overall, Ad4BP/SF-1 potentially maintains the intracellular NADPH level through cooperative regulation of genes involved in the biological processes for consumption and supply.


Adrenal Cortex/metabolism , Aminohydrolases/metabolism , Gene Expression Regulation, Enzymologic , Malate Dehydrogenase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/metabolism , NADP/metabolism , Steroidogenic Factor 1/metabolism , Active Transport, Cell Nucleus , Adrenal Cortex/cytology , Adrenal Cortex/enzymology , Aminohydrolases/genetics , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Malate Dehydrogenase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Multienzyme Complexes/genetics , Mutation , Promoter Regions, Genetic , RNA Interference , Recombinant Fusion Proteins , Steroidogenic Factor 1/antagonists & inhibitors , Steroidogenic Factor 1/genetics
14.
Mol Cell Endocrinol ; 445: 55-64, 2017 04 15.
Article En | MEDLINE | ID: mdl-27940302

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.


Androgens/metabolism , Leydig Cells/cytology , Testis/embryology , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Humans , Leydig Cells/metabolism , Male , Mice , Signal Transduction , Stem Cells/cytology , Testis/cytology , Testis/metabolism
15.
Sex Dev ; 11(2): 53-63, 2017.
Article En | MEDLINE | ID: mdl-27960177

Fetal Leydig cells (FLCs) and adult Leydig cells (ALCs) develop in the mammalian prenatal and postnatal testes, respectively. In mice, FLCs emerge in the interstitial space of the testis as early as embryonic day 12.5 and thereafter increase in number during the fetal stage. We previously established a transgenic mouse line in which FLCs are labeled with EGFP and demonstrated that the EGFP-labeled FLCs were present even in adult testes. However, the characteristics of FLCs during postnatal stages remained unclear. In the present study, a comparison of the transcriptomes of FLCs from prenatal and postnatal testes and of ALCs from adult testes revealed that FLCs gradually alter their characteristics across developmental stages and come to roughly resemble ALCs. Many cholesterogenic genes simultaneously expressed a unique alternation pattern, while many oxidative phosphorylation and ß-oxidation (both mitochondrial functions) genes showed a different unique pattern. These metabolic gene expression alterations might be triggered by milieu changes, such as nutrient and oxygen supply, from the prenatal to the postnatal period.


Aging/genetics , Embryonic Development/genetics , Fetus/cytology , Gene Expression Regulation, Developmental , Leydig Cells/metabolism , Animals , Cholesterol/biosynthesis , Female , Lipid Metabolism/genetics , Male , Mice, Inbred ICR , Organelle Biogenesis , Oxidative Phosphorylation , Steroids/biosynthesis
16.
Endocrinology ; 157(12): 4899-4913, 2016 Dec.
Article En | MEDLINE | ID: mdl-27732090

Adult Leydig cells are derived from proliferating stem/progenitor Leydig cells in the infant testis and subsequent differentiation to steroidogenic cells in adult mice. Leydig cell proliferation in the infant testis occurs primarily in response to increased levels of LH that induce Leydig cell expression of neuregulin 1 (NRG1). Depletion of NRG1 in Nrg1 mutant mice (Nrg1flox;flox;Cyp19a1Cre mice) dramatically reduces Leydig cell proliferation in the infant testes, leading to a reduction of testis weight, epididymial weight, and serum T in the adult mutant mice. The mutant mice are subfertile due to impaired sexual behavior and abnormal elongation of the spermatogenic cells. These defects were reversed by T treatment of the mutant mice in vivo. Furthermore, NRG1 alone induces the proliferation of Leydig cells in cultures of infant (d 10) testes obtained from mutant mice. Collectively these results show that LH induction of NRG1 directly drives the proliferation of Leydig cells in the infant testis, leading to an obligatory number of adult Leydig cells required for the production of sufficient androgen to support and maintain spermatogenesis and sexual behavior of adult male mice.


Cell Proliferation/physiology , Leydig Cells/metabolism , Neuregulin-1/metabolism , Sexual Behavior, Animal/physiology , Spermatogenesis/physiology , Animals , Cell Proliferation/drug effects , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mice , Mice, Knockout , Neuregulin-1/genetics , Sexual Behavior, Animal/drug effects , Spermatogenesis/drug effects , Testosterone/pharmacology
17.
Endocrine ; 54(2): 484-496, 2016 Nov.
Article En | MEDLINE | ID: mdl-27455990

Genetic mutations of steroidogenic factor 1 (also known as Ad4BP or Nr5a1) have increasingly been reported in patients with 46,XY disorders of sex development (46,XY disorders of sex development). However, because the phenotype of 46,XY disorders of sex development with a steroidogenic factor 1 mutation is wide-ranging, its precise diagnosis remains a clinical problem. We previously reported the frequent occurrence of lipid accumulation in Leydig cells among patients with 46,XY disorders of sex development with a steroidogenic factor 1 mutation, an observation also reported by other authors. To address the mechanism of lipid accumulation in this disease, we examined the effects of steroidogenic factor 1 deficiency on downstream targets of steroidogenic factor 1 in in vitro and in vivo. We found that lipid accumulation in Leydig cells was enhanced after puberty in heterozygous steroidogenic factor 1 knockout mice compared with wild-type mice, and was accompanied by a significant decrease in steroidogenic acute regulatory protein and CYP11A1 expression. In mouse Leydig cell lines, steroidogenic factor 1 knockdown induced a remarkable accumulation of neutral lipids and cholesterol with reduced androgen levels. Steroidogenic factor 1 knockdown reduced the expression of steroidogenic acute regulatory protein and CYP11A1, both of which are transcriptional targets of steroidogenic factor 1 and key molecules for steroidogenesis from cholesterol in the mitochondria. Knockdown of either steroidogenic acute regulatory protein or CYP11A1 also induced lipid accumulation, and knockdown of both had an additive effect. Our data suggested that lipid accumulation in the Leydig cells of the 46,XY disorders of sex development phenotype with a steroidogenic factor 1 mutation is due, at least in part, to the suppression of steroidogenic acute regulatory protein and CYP11A1, and a resulting increase in unmetabolized cholesterol.


Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cholesterol/metabolism , Leydig Cells/metabolism , Lipid Metabolism/genetics , Phosphoproteins/metabolism , Steroidogenic Factor 1/genetics , Animals , Male , Mice , Mice, Knockout , Steroidogenic Factor 1/metabolism
18.
Endocrinology ; 157(3): 1222-33, 2016 Mar.
Article En | MEDLINE | ID: mdl-26697723

Fetal and adult Leydig cells develop in mammalian prenatal and postnatal testes, respectively. In mice, fetal Leydig cells (FLCs) emerge in the interstitial space of the testis at embryonic day 12.5 and thereafter increase in number, possibly through differentiation from progenitor cells. However, the progenitor cells have not yet been identified. Previously, we established transgenic mice in which FLCs are labeled strongly with enhanced green fluorescent protein (EGFP). Interestingly, fluorescence-activated cell sorting provided us with weakly EGFP-labeled cells as well as strongly EGFP-labeled FLCs. In vitro reconstruction of fetal testes demonstrated that weakly EGFP-labeled cells contain FLC progenitors. Transcriptome from the 2 cell populations revealed, as expected, marked differences in the expression of genes required for growth factor/receptor signaling and steroidogenesis. In addition, genes for energy metabolisms such as glycolytic pathways and the citrate cycle were activated in strongly EGFP-labeled cells, suggesting that metabolism is activated during FLC differentiation.


Citric Acid Cycle/genetics , Fetus/metabolism , Gene Expression Regulation, Developmental , Glycolysis/genetics , Leydig Cells/metabolism , RNA, Messenger/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Energy Metabolism/genetics , Gene Expression Profiling , Gonadal Steroid Hormones/biosynthesis , Green Fluorescent Proteins/genetics , Immunohistochemistry , Leydig Cells/cytology , Male , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Stem Cells/cytology
19.
Mol Endocrinol ; 29(11): 1581-93, 2015 Nov.
Article En | MEDLINE | ID: mdl-26402718

Two distinct types of Leydig cells emerge during the development of eutherian mammals. Fetal Leydig cells (FLCs) appear shortly after gonadal sex differentiation, and play a crucial role in masculinization of male fetuses. Meanwhile, adult Leydig cells (ALCs) emerge after birth and induce the secondary male-specific sexual maturation by producing testosterone. Previous histological studies suggested that FLCs regress completely soon after birth. Furthermore, gene disruption studies indicated that androgen signaling is dispensable for FLC differentiation but indispensable for postnatal ALC differentiation. Here, we performed lineage tracing of FLCs using a FLC enhancer of the Ad4BP/SF-1 (Nr5a1) gene and found that FLCs persist in the adult testis. Given that postnatal FLCs expressed androgen receptor (AR) as well as LH receptor (LuR), the effects of AR disruption on FLCs and ALCs were analyzed by crossing AR knockout (KO) mice with FLC-specific enhanced green fluorescent protein (EGFP) mice. Moreover, to eliminate the influence of elevated LH levels in ARKO mice, LuRKO mice and AR/LuR double-KO mice were analyzed. The proportion of ALCs to postnatal FLCs was decreased in ARKO mice, and the effect was augmented in the double-KO mice, suggesting that androgen signaling plays important roles in ALCs, but not in FLCs. Finally, ARKO was achieved in an FLC-specific manner (FLCARKO mice), but the FLC number and gene expression pattern appeared unaffected. These findings support the conclusion that FLCs persist as an androgen-independent Leydig subpopulation in the postnatal testis.


Androgens/metabolism , Leydig Cells/metabolism , Receptors, Androgen/genetics , Receptors, LH/genetics , Testis/embryology , Animals , Cell Differentiation/physiology , Cell Lineage , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Androgen/biosynthesis , Receptors, LH/biosynthesis , Signal Transduction , Steroidogenic Factor 1/genetics , Testis/cytology , Testosterone/metabolism
20.
PLoS One ; 10(6): e0128352, 2015.
Article En | MEDLINE | ID: mdl-26039146

It has been established that two developmentally and functionally distinct cell types emerge within the mammalian testis and adrenal gland throughout life. Fetal and adult types of steroidogenic cells (i.e., testicular Leydig cells and adrenocortical cells) develop in the prenatal and postnatal period, respectively. Although the ovary synthesizes steroids postnatally, the presence of fetal-type steroidogenic cells has not been described. We had previously established transgenic mouse lines in which fetal Leydig cells were labeled with an EGFP reporter gene by the FLE (fetal Leydig enhancer) of the Ad4BP/SF-1 (Nr5a1) gene. In the present study, we examined the reporter gene expression in females and found that the reporter gene is turned on in postnatal ovaries. A comparison of the expressions of the EGFP and marker genes revealed that EGFP is expressed in not all but rather a proportion of steroidogenic theca and in interstitial gland cells in the ovary. This finding was further supported by experiments using BAC transgenic mice in which reporter gene expression recapitulated endogenous Ad4BP/SF-1 gene expression. In conclusion, our observations from this study strongly suggest that ovarian theca and interstitial gland cells in mice consist of at least two cell types.


Adrenal Glands/cytology , Cell Lineage/genetics , Leydig Cells/cytology , Pituitary Gland/cytology , Steroidogenic Factor 1/genetics , Theca Cells/cytology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Aging , Animals , Animals, Newborn , Female , Fetus , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Leydig Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Signal Transduction , Steroidogenic Factor 1/metabolism , Theca Cells/classification , Theca Cells/metabolism , Red Fluorescent Protein
...