Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
J Biol Chem ; 289(26): 18152-62, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24828497

ABSTRACT

Regulation of spatiotemporal gene expression in higher eukaryotic cells is critical for the precise and orderly development of undifferentiated progenitors into committed cell types of the adult. It is well known that dynamic epigenomic regulation (including chromatin remodeling and histone modifications by transcriptional coregulator complexes) is involved in transcriptional regulation. Precisely how these coregulator complexes exert their cell type and developing stage-specific activity is largely unknown. In this study we aimed to isolate the histone demethylase lysine-specific demethylase 1 (LSD1) complex from neural cells by biochemical purification. In so doing, we identified myelin transcription factor 1 (MyT1) as a novel LSD1 complex component. MyT1 is a neural cell-specific zinc finger factor, and it forms a stable multiprotein complex with LSD1 through direct interaction. Target gene analysis using microarray and ChIP assays revealed that the Pten gene was directly regulated by the LSD1-MyT1 complex. Knockdown of either LSD1 or MyT1 derepressed the expression of endogenous target genes and inhibited cell proliferation of a neuroblastoma cell line, Neuro2a. We propose that formation of tissue-specific combinations of coregulator complexes is a critical mechanism for tissue-specific transcriptional regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Neurons/enzymology , Oxidoreductases, N-Demethylating/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Histone Demethylases , Histones/metabolism , Mice , Neurons/metabolism , Oxidoreductases, N-Demethylating/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Species Specificity , Transcription Factors/genetics
3.
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
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...