Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36613635

ABSTRACT

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.


Subject(s)
Gonadotrophs , Pituitary Gland , Regulatory Sequences, Nucleic Acid , Steroidogenic Factor 1 , Animals , Male , Mice , Gonadotrophs/metabolism , Introns/genetics , Pituitary Gland/metabolism , Steroidogenic Factor 1/genetics
2.
Sci Rep ; 11(1): 719, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436964

ABSTRACT

The SRY gene induces testis development even in XX individuals. However, XX/Sry testes fail to produce mature sperm, due to the absence of Y chromosome carrying genes essential for spermatogenesis. XX/Sry Sertoli cells show abnormalities in the production of lactate and cholesterol required for germ cell development. Leydig cells are essential for male functions through testosterone production. However, whether XX/Sry adult Leydig cells (XX/Sry ALCs) function normally remains unclear. In this study, the transcriptomes from XY and XX/Sry ALCs demonstrated that immediate early and cholesterogenic gene expressions differed between these cells. Interestingly, cholesterogenic genes were upregulated in XX/Sry ALCs, although downregulated in XX/Sry Sertoli cells. Among the steroidogenic enzymes, CYP17A1 mediates steroid 17α-hydroxylation and 17,20-lyase reaction, necessary for testosterone production. In XX/Sry ALCs, the latter reaction was selectively decreased. The defects in XX/Sry ALCs, together with those in the germ and Sertoli cells, might explain the infertility of XX/Sry testes.


Subject(s)
Disorders of Sex Development/pathology , Gene Expression Regulation, Developmental , Leydig Cells/pathology , Sertoli Cells/pathology , Sex-Determining Region Y Protein/metabolism , Spermatogenesis , Testis/pathology , Animals , Disorders of Sex Development/genetics , Disorders of Sex Development/metabolism , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sertoli Cells/metabolism , Sex Differentiation , Sex-Determining Region Y Protein/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Testis/metabolism , X Chromosome , Y Chromosome
3.
Development ; 145(23)2018 12 05.
Article in English | MEDLINE | ID: mdl-30518625

ABSTRACT

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.


Subject(s)
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
4.
Commun Biol ; 1: 18, 2018.
Article in English | MEDLINE | ID: mdl-30271905

ABSTRACT

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.

5.
Sci Rep ; 7: 41912, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28150810

ABSTRACT

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.


Subject(s)
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
6.
Endocr J ; 64(3): 315-324, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28202838

ABSTRACT

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.


Subject(s)
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
7.
Sex Dev ; 11(2): 53-63, 2017.
Article in English | MEDLINE | ID: mdl-27960177

ABSTRACT

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.


Subject(s)
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
8.
Endocrinology ; 157(3): 1222-33, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26697723

ABSTRACT

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.


Subject(s)
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
9.
Mol Endocrinol ; 29(11): 1581-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26402718

ABSTRACT

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.


Subject(s)
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
10.
PLoS One ; 10(6): e0128352, 2015.
Article in English | MEDLINE | ID: mdl-26039146

ABSTRACT

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.


Subject(s)
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
11.
Nat Commun ; 5: 3634, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24727981

ABSTRACT

Genetic deficiencies in transcription factors can lead to the loss of certain types of cells and tissue. The steroidogenic tissue-specific nuclear receptor Ad4BP/SF-1 (NR5A1) is one such gene, because mice in which this gene is disrupted fail to develop the adrenal gland and gonads. However, the specific role of Ad4BP/SF-1 in these biological events remains unclear. Here we use chromatin immunoprecipitation sequencing to show that nearly all genes in the glycolytic pathway are regulated by Ad4BP/SF-1. Suppression of Ad4BP/SF-1 by small interfering RNA reduces production of the energy carriers ATP and nicotinamide adenine dinucleotide phosphate, as well as lowers expression of genes involved in glucose metabolism. Together, these observations may explain tissue dysgenesis as a result of Ad4BP/SF-1 gene disruption in vivo. Considering the function of estrogen-related receptor α, the present study raises the possibility that certain types of nuclear receptors regulate sets of genes involved in metabolic pathways to generate energy carriers.


Subject(s)
Steroidogenic Factor 1/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Glucose/metabolism , Glycolysis/genetics , Glycolysis/physiology , Humans , Mice , NADP/metabolism , Pregnenolone/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1/genetics
12.
PLoS One ; 8(6): e68050, 2013.
Article in English | MEDLINE | ID: mdl-23840809

ABSTRACT

Development of the testis begins with the expression of the SRY gene in pre-Sertoli cells. Soon after, testis cords containing Sertoli and germ cells are formed and fetal Leydig cells subsequently develop in the interstitial space. Studies using knockout mice have indicated that multiple genes encoding growth factors and transcription factors are implicated in fetal Leydig cell differentiation. Previously, we demonstrated that the Arx gene is implicated in this process. However, how ARX regulates Leydig cell differentiation remained unknown. In this study, we examined Arx KO testes and revealed that fetal Leydig cell numbers largely decrease throughout the fetal life. Since our study shows that fetal Leydig cells rarely proliferate, this decrease in the KO testes is thought to be due to defects of fetal Leydig progenitor cells. In sexually indifferent fetal gonads of wild type, ARX was expressed in the coelomic epithelial cells and cells underneath the epithelium as well as cells at the gonad-mesonephros border, both of which have been described to contain progenitors of fetal Leydig cells. After testis differentiation, ARX was expressed in a large population of the interstitial cells but not in fetal Leydig cells, raising the possibility that ARX-positive cells contain fetal Leydig progenitor cells. When examining marker gene expression, we observed cells as if they were differentiating into fetal Leydig cells from the progenitor cells. Based on these results, we propose that ARX acts as a positive factor for differentiation of fetal Leydig cells through functioning at the progenitor stage.


Subject(s)
Cell Differentiation/genetics , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Leydig Cells/physiology , Sex Differentiation/genetics , Stem Cells/pathology , Transcription Factors/genetics , Animals , Cell Proliferation , Epithelial Cells/physiology , Epithelium/physiology , Fetus/physiology , Germ Cells/growth & development , Germ Cells/physiology , Gonads/growth & development , Gonads/physiology , Male , Mesonephros/growth & development , Mesonephros/physiology , Mice , Mice, Inbred ICR , Mice, Knockout , Testis/growth & development , Testis/physiology
13.
Mol Endocrinol ; 27(1): 63-73, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23125070

ABSTRACT

Testosterone is a final product of androgenic hormone biosynthesis, and Leydig cells are known to be the primary source of androgens. In the mammalian testis, two distinct populations of Leydig cells, the fetal and the adult Leydig cells, develop sequentially, and these two cell types differ both morphologically and functionally. It is well known that the adult Leydig cells maintain male reproductive function by producing testosterone. However, it has been controversial whether fetal Leydig cells can produce testosterone, and the synthetic pathway of testosterone in the fetal testis is not fully understood. In the present study, we generated transgenic mice in which enhanced green fluorescence protein was expressed under the control of a fetal Leydig cell-specific enhancer of the Ad4BP/SF-1 (Nr5a1) gene. The transgene construct was prepared by mutating the LIM homeodomain transcription factor (LHX9)-binding sequence in the promoter, which abolished promoter activity in the undifferentiated testicular cells. These transgenic mice were used to collect highly pure fetal Leydig cells. Gene expression and steroidogenic enzyme activities in the fetal Leydig cells as well as in the fetal Sertoli cells and adult Leydig cells were analyzed. Our results revealed that the fetal Leydig cells synthesize only androstenedione because they lack expression of Hsd17b3, and fetal Sertoli cells convert androstenedione to testosterone, whereas adult Leydig cells synthesize testosterone by themselves. The current study demonstrated that both Leydig and Sertoli cells are required for testosterone synthesis in the mouse fetal testis.


Subject(s)
Leydig Cells/metabolism , Sertoli Cells/metabolism , Testosterone/biosynthesis , Animals , Base Sequence , Binding Sites , Cells, Cultured , Conserved Sequence , Fetus/cytology , Gene Expression , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Steroidogenic Factor 1/genetics , Testis/cytology , Testis/metabolism
14.
Endocrinology ; 153(2): 913-24, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22186409

ABSTRACT

Mice lacking the function of the polycomb group protein CBX2 (chromobox homolog 2; also known as M33) show defects in gonadal, adrenal, and splenic development. In particular, XY knockout (KO) mice develop ovaries but not testes, and the gonads are hypoplastic in both sexes. However, how CBX2 regulates development of these tissues remains largely unknown. In the present study, we used microarray, RT-PCR, and immunohistochemical analyses to show that the expression of Sry, Sox9, Lhx9, Ad4BP/SF-1, Dax-1, Gata4, Arx, and Dmrt1, genes encoding transcription factors essential for gonadal development, is affected in Cbx2 KO gonads. Male-to-female sex reversal in Cbx2 KO mice was rescued by crossing them with transgenic mice displaying forced expression of Sry or Sox9. However, testes remained hypoplastic in these mice, indicating that the size and the sex of the gonad are determined by different sets of genes. Our study implicates Cbx2 in testis differentiation through regulating Sry gene expression.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Repressor Proteins/metabolism , Sex-Determining Region Y Protein/metabolism , Testis/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Array Analysis , Repressor Proteins/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sex Determination Processes/physiology , Sex-Determining Region Y Protein/genetics , Testis/cytology , Testis/growth & development
15.
Endocrinology ; 153(1): 417-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22128023

ABSTRACT

Adrenal 4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1) (Nr5a1) is a nuclear receptor essential for reproductive tissue development and endocrine regulation. This factor is expressed in steroidogenic tissues (e.g. adrenal glands and gonads), and expression of this factor is tightly regulated in a tissue and cell type-specific manner. Our previous studies have identified tissue and cell type-specific enhancers in the introns of the Ad4BP/SF-1 gene in fetal adrenal glands, ventromedial hypothalamus, and pituitary gonadotrope. Characterization of the enhancers had provided new insights into tissue and cell development. However, these studies have failed to identify any gonad-specific enhancer. Here, we identified a fetal Leydig cell-specific enhancer in the upstream region of the mouse Ad4BP/SF-1 gene using transgenic mouse assays. Alignment of the upstream regions among vertebrate animal species demonstrated that the enhancer consisted of three conserved regions, whereby the most highly conserved region contained an Ad4BP/SF-1 binding sequence and an E-box. Mutation of each sequence abolished the enhancer activity and led to a loss of reporter gene expression. These results suggested that Ad4BP/SF-1 gene expression in the fetal Leydig cell is regulated by a yet unidentified E-box binding protein(s) and by an autoregulatory loop formed by Ad4BP/SF-1. Although fetal Leydig cells have been thought to play crucial roles for masculinization of various fetal tissues through androgen production, other functions have remained elusive. Our identification of a fetal Leydig cell-specific enhancer in the Ad4BP/SF-1 gene would be a powerful tool to address these gaps in the knowledge base.


Subject(s)
Enhancer Elements, Genetic , Leydig Cells/metabolism , Steroidogenic Factor 1/genetics , Animals , Base Sequence , Binding Sites/genetics , Conserved Sequence , DNA Primers/genetics , E-Box Elements , Fetus/cytology , Fetus/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Lac Operon , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Species Specificity
16.
Endocrinology ; 151(12): 5893-904, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20962046

ABSTRACT

The gonadal primordium first emerges as a thickening of the embryonic coelomic epithelium, which has been thought to migrate mediodorsally to form the primitive gonad. However, the early gonadal development remains poorly understood. Mice lacking the paired-like homeobox gene Emx2 display gonadal dysgenesis. Interestingly, the knockout (KO) embryonic gonads develop an unusual surface accompanied by aberrant tight junction assembly. Morphological and in vitro cell fate mapping studies showed an apparent decrease in the number of the gonadal epithelial cells migrated to mesenchymal compartment in the KO, suggesting that polarized cell division and subsequent cell migration are affected. Microarray analyses of the epithelial cells revealed significant up-regulation of Egfr in the KO, indicating that Emx2 suppresses Egfr gene expression. This genetic correlation between the two genes was reproduced with cultured M15 cells derived from mesonephric epithelial cells. Epidermal growth factor receptor signaling was recently shown to regulate tight junction assembly through sarcoma viral oncogene homolog tyrosine phosphorylation. We show through Emx2 KO analyses that sarcoma viral oncogene homolog tyrosine phosphorylation, epidermal growth factor receptor tyrosine phosphorylation, and Egfr expression are up-regulated in the embryonic gonad. Our results strongly suggest that Emx2 is required for regulation of tight junction assembly and allowing migration of the gonadal epithelia to the mesenchyme, which are possibly mediated by suppression of Egfr expression.


Subject(s)
Epithelial Cells/cytology , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental/physiology , Gonads/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Cell Proliferation , ErbB Receptors/genetics , Gene Expression Profiling , Gonads/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Protein Array Analysis , Tight Junctions/physiology , Transcription Factors/genetics
17.
Histol Histopathol ; 23(11): 1387-98, 2008 11.
Article in English | MEDLINE | ID: mdl-18785121

ABSTRACT

The development of mature ovarian follicles is greatly dependent on healthy thecal angiogenesis. Recent experimental evidence showed that thyroxine (T4) treatment promoted ovarian follicle development in immature hypothyroid (rdw) rats. However, an involvement of thyroid hormone in ovarian follicular angiogenesis has not yet been demonstrated. By morphological and molecular approaches, the present studies demonstrated that antral follicles in untreated, T4- or equine chorionic gonadotropin (eCG)-treated rdw rats were mainly small and/or atretic, and presented a poorly developed thecal microvasculature with ultrastructural evidence of diffuse quiescent or degenerative thin capillaries. However, T4 together with eCG increased the number of large antral and mature follicles with numerous activated capillaries and ultra-structural evidence of rich and diffuse angiogenesis in the theca layer. While T4 alone significantly increased mRNA expression of vascular endothelial growth factor (VEGF) and tumor necrosis factor alpha (TNFalpha), it decreased that of fetal liver kinase compared with those in the untreated group. Combined treatment of T4 and eCG markedly increased mRNA abundance of not only VEGF and TNFalpha, but also basic fibroblast growth factor. These data suggest that T4 may promote ovarian follicular angiogenesis in rdw rats by up-regulating mRNA expression of major angiogenic factors.


Subject(s)
Angiogenic Proteins/metabolism , Chorionic Gonadotropin/pharmacology , Hypothyroidism/drug therapy , Neovascularization, Physiologic/drug effects , Ovarian Follicle/blood supply , Thyroxine/pharmacology , Angiogenic Proteins/genetics , Animals , Corrosion Casting , Disease Models, Animal , Female , Fibroblast Growth Factor 2/metabolism , Hypothyroidism/metabolism , Hypothyroidism/pathology , Hypothyroidism/physiopathology , Microcirculation/drug effects , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
18.
Mol Reprod Dev ; 75(9): 1361-71, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18288644

ABSTRACT

Forkhead transcription factors are characterized by a winged helix DNA binding domain, and the members of this family are classified into 20 subclasses by phylogenetic analyses. Fkhl18 is structurally unique, and is classified into FoxS subfamily. We found Fkhl18 expression in periendothelial cells of the developing mouse fetal testis. In an attempt to clarify its function, we generated mice with Fkhl18 gene disruption. Although KO mice developed normally and were fertile in both sexes, we frequently noticed unusual blood accumulation in the fetal testis. Electron microscopic analysis demonstrated frequent gaps, measuring 100-400 nm, in endothelial cells of blood vessels. These gaps probably represented ectopic apoptosis of testicular periendothelial cells, identified by caspase-3 expression, in KO fetuses. No apoptosis of endothelial cells was noted. Fkhl18 suppressed the transcriptional activity of FoxO3a and FoxO4. Considering that Fas ligand gene expression is activated by Foxs, the elevated activity of Foxs in the absence of Fkhl18 probably explains the marked apoptosis of periendothelial cells in Fkhl18 KO mice.


Subject(s)
Neovascularization, Physiologic/genetics , Testis/embryology , Transcription Factors/physiology , Animals , Apoptosis/genetics , Blood Vessels/abnormalities , Blood Vessels/embryology , Blood Vessels/metabolism , Embryo, Mammalian , Endothelium, Vascular/metabolism , Fas Ligand Protein/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/physiology , Gene Expression Regulation, Developmental , Lac Operon , Male , Mice , Mice, Transgenic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Testis/blood supply , Testis/metabolism , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Reproduction ; 134(5): 677-82, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17965258

ABSTRACT

Vascular endothelial growth factor (VEGF) expression in granulosa cells is associated with the thecal vasculature growth during ovarian follicular development. We hypothesized that injection of VEGF gene fragments directly into the rat ovary would induce production of a large number of ovulatory follicles and that these follicles would ovulate. To test this hypothesis, we treated immature female rats with combinations of hormones and VEGF gene fragments. The animals were divided into two groups: one group received solution containing transfection reagents as a control (n = 5), while the other group received direct ovarian injection of VEGF gene fragments at 19 (n = 5), 21 (n = 5), 23 (n = 5), or 25 (n = 5) days after birth followed by i.p. administration of 20 IU equine chorionic gonadotropin (eCG) at the age of 26 days. Forty-eight hours after eCG injection, animals were given 20 IU human chorionic gonadotropin (hCG) i.p. and then the oocytes in both groups were counted. The maximum number of ovulated oocytes was obtained when the VEGF gene fragments were injected into the rat ovary at 21 days after birth. Histological examination revealed that the injection of VEGF gene fragments markedly increased the vascular density around the preovulatory follicles and also the number of these follicles. Our data provide the first reported evidence that most ovulatory follicles generated by injection of VEGF gene fragments are able to ovulate upon hCG treatment. These results demonstrate that injection of VEGF gene fragments directly into the ovary stimulates the development of antral follicles by inducing the formation of thecal vasculature in immature female rats.


Subject(s)
Ovarian Follicle/growth & development , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Animals , Capillaries , Female , Gene Expression , Gonadotropins, Equine/pharmacology , Injections , Organ Size , Ovarian Follicle/blood supply , Ovarian Follicle/drug effects , Ovary/anatomy & histology , Ovulation/drug effects , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stimulation, Chemical , Theca Cells/cytology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
20.
Nat Genet ; 38(12): 1369-71, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086185

ABSTRACT

46,XY disorders of sex development (DSD) refer to a wide range of abnormal genitalia, including hypospadias, which affects approximately 0.5% of male newborns. We identified three different nonsense mutations of CXorf6 in individuals with hypospadias and found that its mouse homolog was specifically expressed in fetal Sertoli and Leydig cells around the critical period for sex development. These data imply that CXorf6 is a causative gene for hypospadias.


Subject(s)
Chromosomes, Human, X/genetics , Hypospadias/genetics , Animals , Base Sequence , Codon, Nonsense , DNA/genetics , Female , Gene Expression Regulation, Developmental , Humans , Hypospadias/embryology , In Situ Hybridization , Infant, Newborn , Male , Mice , Open Reading Frames , Pedigree , Pregnancy , Sex Differentiation/genetics , Testis/abnormalities , Testis/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...