Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Clin Exp Hypertens ; 39(3): 257-263, 2017.
Article in English | MEDLINE | ID: mdl-28448185

ABSTRACT

This study was designed to assess the effect of additional spironolactone on blood pressure in patients with resistant hypertension. MEDLINE, EMBASE, and Cochrane Library were searched to identify randomized controlled trials (RCTs) that determined the effect of add-on spironolactone on blood pressure in patients with resistant hypertension compared with a control group. A total of five RCTs met the inclusion criteria. Spironolactone reduced office systolic blood pressure (SBP) by 15.73 mmHg (95% CI -20.45 to -11.0; P < 0.00001) and office diastolic blood pressure (DBP) by 6.21 mmHg (95% CI -8.33 to -4.1, P < 0.00001) as compared to placebo group. The pooled changes of 24 h ambulatory or home SBP and DBP were -8.7 mmHg (95% CI -8.79 to -8.62, P < 0.00001) and -4.12 mmHg (95% CI -4.48 to -3.75, P < 0.00001), in favor of the spironolactone group. In comparison with alternative drugs including beta-blocker, candesartan, or alpha methyldopa, spironolactone reduced home SBP by 4.5 mmHg (95% CI -4.63 to -4.37, P < 0.00001). Addition of spironolactone provides benefit effect on blood pressure in patients with resistant hypertension.


Subject(s)
Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Spironolactone/therapeutic use , Drug Resistance , Drug Therapy, Combination , Humans , Randomized Controlled Trials as Topic , Spironolactone/pharmacology
2.
Biochim Biophys Acta ; 1839(1): 33-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24321386

ABSTRACT

Ferredoxin reductase (FDXR, also known as adrenodoxin reductase) is a mitochondrial flavoprotein that transfers electrons from NADPH to mitochondrial cytochrome P450 enzymes, mediating the function of an iron-sulfur cluster protein, ferredoxin. FDXR functions in various metabolic processes including steroidogenesis. It is well known that multiple steroidogenic enzymes are regulated by a transcription factor steroidogenic factor-1 (SF-1, also known as Ad4BP). Previously, we have shown that SF-1 transduction causes human mesenchymal stem cell differentiation into steroidogenic cells. Genome-wide analysis of differentiated cells, using a combination of DNA microarray and promoter tiling array analyses, showed that FDXR is a novel SF-1 target gene. In this study, the transcriptional regulatory mechanism of FDXR was examined in steroidogenic cells. A chromatin immunoprecipitation assay revealed that a novel SF-1 binding region was located within intron 2 of the human FDXR gene. Luciferase reporter assays showed that FDXR transcription was activated through the novel SF-1 binding site within intron 2. Endogenous SF-1 knockdown in human adrenocortical H295R and KGN cells decreased FDXR expression. In H295R cells, strong binding of two histone markers of active enhancers, histones H3K27ac and H3K4me2, were detected near the SF-1 binding site within intron 2. Furthermore, the binding of these histone markers was decreased concurrent with SF-1 knockdown in H295R cells. These results indicated that abundant FDXR expression in these steroidogenic cells was maintained through SF-1 binding to the intronic enhancer of the FDXR gene.


Subject(s)
Enhancer Elements, Genetic , Ferredoxin-NADP Reductase/genetics , Steroidogenic Factor 1/genetics , Steroids/metabolism , Transcription, Genetic , Binding Sites , Cell Line , DNA-Binding Proteins , Ferredoxin-NADP Reductase/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Histones/genetics , Humans , Introns , Jumonji Domain-Containing Histone Demethylases/genetics , Regulatory Sequences, Nucleic Acid , Steroidogenic Factor 1/metabolism , Steroids/biosynthesis
3.
Biochim Biophys Acta ; 1839(5): 406-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24705138

ABSTRACT

Pluripotent stem cells maintain their pluripotency and undifferentiated status through a network of transcription factors. Liver receptor homolog-1 (Lrh-1) is one of these, and regulates the expression of other important transcription factors such as Oct-3/4 and Nanog. In early embryo and embryonic stem (ES) cells, Lrh-1 is transcribed using a unique promoter. In this study, we investigated the transcriptional regulation of embryonic Lrh-1 using ES and embryonal carcinoma F9 cells. Reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays demonstrated that Sox2 and Gabp proteins bind to the promoter region of embryonic Lrh-1, and are necessary for its activation. The Sox2 site showed strong promoter activity and affinity for protein binding. Upon differentiation into the parietal endoderm by retinoic acid and cAMP, Lrh-1 promoter activity and transcripts were markedly reduced within 24 h. At the same time, Sox2 and Gabp binding to the promoter region of Lrh-1 were decreased, followed by a reduction of their expression. These results indicate that embryonic Lrh-1 expression is regulated by both Sox2 and Gabp. Our study presents new insights into the transcription factor network of pluripotent stem cells.


Subject(s)
Embryonic Stem Cells/physiology , GA-Binding Protein Transcription Factor/genetics , Receptors, Cytoplasmic and Nuclear/genetics , SOXB1 Transcription Factors/genetics , Animals , Base Sequence , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , GA-Binding Protein Transcription Factor/metabolism , Gene Expression Regulation , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Promoter Regions, Genetic , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , SOXB1 Transcription Factors/metabolism , Transcription, Genetic , Transfection
4.
Cell Physiol Biochem ; 37(4): 1619-28, 2015.
Article in English | MEDLINE | ID: mdl-26536571

ABSTRACT

BACKGROUND/AIMS: Cardiac malfunction is a common complication in sepsis and significantly increases the mortality of patients in septic shock. However, no studies have examined whether andrographolide (And) reduces LPS-induced myocardial malfunction. METHODS: Left ventricular systolic and diastolic functions were examined using echocardiography. TNF-α and IL-1ß protein levels were detected by an enzyme-linked immunosorbent assay (ELISA). NO oxidation products were determined using Griess reagent. Protein expression levels of inhibitors of NF-κBα (IκB) and phospho-IκB were determined via Western blot. Oxidative injury was determined by measuring myocardial lipid peroxidation and superoxide dismutase activity. Cardiac apoptosis was examined by terminal deoxynucleotidyl transferase-mediated dUTP nickend-labeling (TUNEL) and cardiac caspase 3/7 activity. RESULTS: And blunted LPS-induced myocardial malfunctions in mice. LPS induced TNF-α, IL-1ß, and NO production as well as I-κB phosphorylation. Cardiac apoptosis was attenuated via incubation with And, but the extent of oxidative injury remained unaffected. CONCLUSION: And prevents LPS-induced cardiac malfunctions in mice by inhibiting TNF-α, IL-1ß, and NO production, IκB phosphorylation, and cardiac apoptosis, indicating that And may be a potential agent for preventing myocardial malfunction during sepsis.


Subject(s)
Apoptosis/drug effects , Diterpenes/pharmacology , Heart/drug effects , I-kappa B Proteins/metabolism , Myocardium/metabolism , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Echocardiography , Interleukin-1beta/metabolism , Lipid Peroxidation/drug effects , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Phosphorylation/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ventricular Function, Left/drug effects
5.
Biochem J ; 460(3): 459-71, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24635384

ABSTRACT

The transcription factor SF-1 (steroidogenic factor-1) is a master regulator of steroidogenesis. Previously, we have found that SF-1 induces the differentiation of mesenchymal stem cells into steroidogenic cells. To elucidate the molecular mechanisms of SF-1-mediated functions, we attempted to identify protein components of the SF-1 nuclear protein complex in differentiated cells. SF-1 immunoaffinity chromatography followed by MS/MS analysis was performed, and 24 proteins were identified. Among these proteins, we focused on C/EBPß (CCAAT/enhancer-binding protein ß), which is an essential transcription factor for ovulation and luteinization, as the transcriptional mechanisms of C/EBPß working together with SF-1 are poorly understood. C/EBPß knockdown attenuated cAMP-induced progesterone production in granulosa tumour-derived KGN cells by altering STAR (steroidogenic acute regulatory protein), CYP11A1 (cytochrome P450, family 11, subfamily A, polypeptide 1) and HSD3B2 (hydroxy-δ-5-steroid dehydrogenase, 3ß- and steroid δ-isomerase 2) expression. EMSA and ChIP assays revealed novel C/EBPß-binding sites in the upstream regions of the HSD3B2 and CYP11A1 genes. These interactions were enhanced by cAMP stimulation. Luciferase assays showed that C/EBPß-responsive regions were found in each promoter and C/EBPß is involved in the cAMP-induced transcriptional activity of these genes together with SF-1. These results indicate that C/EBPß is an important mediator of progesterone production by working together with SF-1, especially under tropic hormone-stimulated conditions.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Progesterone/biosynthesis , Steroidogenic Factor 1/physiology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Gene Expression Regulation , Humans , Mice , Phosphoproteins , Progesterone/genetics , Progesterone Reductase/genetics , Tandem Mass Spectrometry
6.
FASEB J ; 27(8): 3198-208, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23650189

ABSTRACT

Steroidogenic factor 1 (SF-1) is a master regulator for steroidogenesis. In this study, we identified novel SF-1 target genes using a genome-wide promoter tiling array and a DNA microarray. SF-1 was found to regulate human glutathione S-transferase A (GSTA) family genes (hGSTA1-hGSTA4), a superfamily of detoxification enzymes clustered on chromosome 6p12. All hGSTA genes were up-regulated by transduction of SF-1 into human mesenchymal stem cells, while knockdown of endogenous SF-1 in H295R cells down-regulated all hGSTA genes. Chromatin immunoprecipitation assays, however, revealed that SF-1 bound directly to the promoters of hGSTA3 and weakly of hGSTA4. Chromosome conformation capture assays revealed that the coordinated expression of the genes was based on changes in higher-order chromatin structure triggered by SF-1, which enables the formation of long-range interactions, at least between hGSTA1 and hGSTA3 gene promoters. In steroidogenesis, dehydrogenation of the 3-hydroxy group and subsequent Δ(5)-Δ(4) isomerization are thought to be enzymatic properties of 3ß-hydroxysteroid dehydrogenase (3ß-HSD). Here, we demonstrated that, in steroidogenic cells, the hGSTA1 and hGSTA3 gene products catalyze Δ(5)-Δ(4) isomerization in a coordinated fashion with 3ß-HSD II to produce progesterone or Δ(4)-androstenedione from their Δ(5)-precursors. Thus, hGSTA1 and hGSTA3 gene products are new members of steroidogenesis working as Δ(5)-Δ(4) isomerases.


Subject(s)
Glutathione Transferase/metabolism , Isoenzymes/metabolism , Steroidogenic Factor 1/metabolism , Steroids/biosynthesis , Androstenedione/biosynthesis , Blotting, Western , Cell Line , Cell Line, Tumor , Gene Expression Regulation , Glutathione Transferase/chemical synthesis , Glutathione Transferase/genetics , Humans , Isoenzymes/genetics , Mesenchymal Stem Cells/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Progesterone/biosynthesis , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1/genetics
7.
J Biol Chem ; 285(36): 28240-51, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20601698

ABSTRACT

StAR (steroidogenic acute regulatory protein) mediates the transport of cholesterol from the outer to the inner mitochondrial membrane, the process of which is the rate-limiting step for steroidogenesis. Transcriptional regulation of the proximal promoter of the human StAR gene has been well characterized, whereas analysis of its distal control region has not. Recently, we found that SF-1 (steroidogenic factor 1) induced the differentiation of mesenchymal stem cells (MSCs) into steroidogenic cells with the concomitant strong induction of StAR expression. Here, we show, using differentiated MSCs, that StAR expression is regulated by a novel distal control region. Using electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays, we identified novel SF-1 binding sites between 3,000 and 3,400 bp upstream of StAR. A luciferase reporter assay revealed that the region worked as a strong regulator to exert maximal transcription of StAR. ChIP analysis of histone H3 revealed that upon SF-1 expression, nucleosome eviction took place at the SF-1 binding sites, not only in the promoter but also in the distal SF-1 binding sites. Chromosome conformation capture analysis revealed that the region upstream of StAR formed a chromatin loop both in the differentiated MSCs and in KGN cells, a human granulosa cell tumor cell line, where SF-1 is endogenously expressed. Finally, SF-1 knockdown resulted in disrupted formation of this chromatin loop in KGN cells. These results indicate that the novel distal control region participate in StAR activation through SF-1 dependent alterations of chromatin structure, including histone eviction and chromatin loop formation.


Subject(s)
Chromatin/metabolism , Locus Control Region/genetics , Phosphoproteins/genetics , Steroidogenic Factor 1/metabolism , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation , Conserved Sequence , Electrophoretic Mobility Shift Assay , Evolution, Molecular , Histones/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mice , Molecular Sequence Data , Phosphoproteins/metabolism , Rats , Transcriptional Activation
8.
Mol Cell Endocrinol ; 370(1-2): 1-10, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23435367

ABSTRACT

Ferredoxin 1 (FDX1; adrenodoxin) is an iron-sulfur protein that is involved in various metabolic processes, including steroid hormone synthesis in mammalian tissues. We investigated the transcriptional regulation of FDX1 in ovarian granulosa cells. Previously, we reported that the NR5A family, including steroidogenic factor-1 (SF-1) and liver receptor homolog-1 could induce differentiation of human mesenchymal stem cells (hMSCs) into steroidogenic cells. A ChIP assay showed that SF-1 could bind to the FDX1 promoter in differentiated hMSCs. Luciferase reporter assays showed that transcription of FDX1 was synergistically activated by the NR5A family and 8Br-cAMP treatment through two SF-1 binding sites and a CRE-like sequence in a human ovarian granulosa cell line, KGN. Knockdown of FDX1 attenuated progesterone production in KGN cells. These results indicate transcription of FDX1 is regulated by the NR5A family and cAMP signaling, and participates in steroid hormone production in ovarian granulosa cells.


Subject(s)
Ferredoxins/genetics , Granulosa Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adrenodoxin/genetics , Animals , Cell Differentiation , Cell Line, Tumor , Cyclic AMP/metabolism , DNA-Binding Proteins/metabolism , Female , Ferredoxins/biosynthesis , Gene Expression Regulation , HeLa Cells , Humans , Mesenchymal Stem Cells/metabolism , Progesterone/biosynthesis , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering , Rats , Rats, Wistar , Signal Transduction , Steroidogenic Factor 1/drug effects , Transcription, Genetic
9.
Endocrinology ; 154(4): 1648-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23471216

ABSTRACT

Liver receptor homolog-1 (LRH-1) is a member of the nuclear receptor 5A (NR5A) subfamily. It is expressed in granulosa cells of the ovary and is involved in steroidogenesis and ovulation. To reveal the transcriptional regulatory mechanism of LRH-1, we determined its transcription start site in the ovary using KGN cells, a human granulosa cell tumor cell line. 5'-rapid amplification of cDNA ends PCR revealed that human ovarian LRH-1 was transcribed from a novel transcription start site, termed exon 2o, located 41 bp upstream of the reported exon 2. The novel LRH-1 isoform was expressed in the human ovary but not the liver. Promoter analysis and an EMSA indicated that a steroidogenic factor-1 (SF-1) binding site and a GC box upstream of exon 2o were required for promoter activity, and that SF-1 and specificity protein (Sp)-1/3 bind to the respective regions in ovarian granulosa cells. In KGN cells, transfection of SF-1 increased ovarian LRH-1 promoter activity and SF-1-dependent reporter activity was further enhanced when peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) was cotransfected. In Drosophila SL2 cells, Sp1 was more effective than Sp3 in enhancing promoter activity, and co-transfection of the NR5A-family synergistically increased activity. Infection with adenoviruses expressing SF-1 or PGC-1α induced LRH-1 expression in KGN cells. These results indicate that the expression of human LRH-1 is regulated in a tissue-specific manner, and that the novel promoter region is controlled by the Sp-family, NR5A-family and PGC-1α in ovarian granulosa cells in a coordinated fashion.


Subject(s)
Granulosa Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Steroidogenic Factor 1/metabolism , Animals , Cell Line , Cell Line, Tumor , Drosophila , Female , Gene Expression Regulation , Heat-Shock Proteins/metabolism , Humans , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Transcription Factors/metabolism
10.
Mol Cell Endocrinol ; 369(1-2): 42-51, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23415714

ABSTRACT

It is well known that the androgen/androgen receptor (AR) pathway is involved in both male and female fertility in mammals. AR knockout female mice are reported to exhibit various abnormalities in follicle development, and a subfertile phenotype. In exogenous gonadotropin-induced superovulation, serum androgen levels were robustly elevated in female mice at the periovulatory stage after human chorionic gonadotropin (hCG) treatment. At this stage, ovarian AR proteins were strongly expressed in cumulus cells. Because these results suggested that the androgen/AR pathway is involved in ovulation, we investigated the expression of ovulation-related genes in the mouse ovary treated with the nonaromatizable androgen, 5α-dihydrotestosterone (DHT). DHT treatment induced the expression of the genes for cyclooxyganase-2 (Cox-2 or prostaglandin endoperoxidase synthase 2) and the epidermal growth factor-like factor, amphiregulin (Areg), in the ovary, whereas their hCG-induced expression was suppressed by the AR antagonist flutamide. These genes were also induced by DHT in AR-expressing primary granulosa and granulosa tumor-derived cells. Reporter assays, electrophoretic shift mobility assays and chromatin immunoprecipitation assays demonstrated that androgen response sequence(s) existing upstream of each gene were responsible for androgen responsiveness and were occupied by the AR in periovulatory granulosa cells. Our results suggest that the androgen/AR pathway is involved in the ovulatory process via expression of the Cox-2 and Areg genes in periovulatory granulosa cells.


Subject(s)
Androgens/pharmacology , Cyclooxygenase 2/metabolism , Dihydrotestosterone/pharmacology , Glycoproteins/metabolism , Granulosa Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Amphiregulin , Androgen Receptor Antagonists/pharmacology , Animals , Chorionic Gonadotropin/pharmacology , Cyclooxygenase 2/genetics , EGF Family of Proteins , Female , Flutamide/pharmacology , Glycoproteins/genetics , Granulosa Cells/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL
11.
Endocrinology ; 153(11): 5522-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23024262

ABSTRACT

5-Aminolevulinic acid synthase 1 (ALAS1) is a rate-limiting enzyme for heme biosynthesis in mammals. Heme is essential for the catalytic activities of P450 enzymes including steroid metabolic enzymes. Nuclear receptor 5A (NR5A) family proteins, steroidogenic factor-1 (SF-1), and liver receptor homolog-1 (LRH-1) play pivotal roles in regulation of steroidogenic enzymes. Recently, we showed that expression of SF-1/LRH-1 induces differentiation of mesenchymal stem cells into steroidogenic cells. In this study, genome-wide analysis revealed that ALAS1 was a novel SF-1-target gene in differentiated mesenchymal stem cells. Chromatin immunoprecipitation and reporter assays revealed that SF-1/LRH-1 up-regulated ALAS1 gene transcription in steroidogenic cells via binding to a 3.5-kb upstream region of ALAS1. The ALAS1 gene was up-regulated by overexpression of SF-1/LRH-1 in steroidogenic cells and down-regulated by knockdown of SF-1 in these cells. Peroxisome proliferator-activated receptor-γ coactivator-1α, a coactivator of nuclear receptors, also strongly coactivated expression of NR5A-target genes. Reporter analysis revealed that peroxisome proliferator-activated receptor-γ coactivator-1α strongly augmented ALAS1 gene transcription caused by SF-1 binding to the 3.5-kb upstream region. Finally knockdown of ALAS1 resulted in reduced progesterone production by steroidogenic cells. These results indicate that ALAS1 is a novel NR5A-target gene and participates in steroid hormone production.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Granulosa Cells/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Steroidogenic Factor 1/genetics , 5-Aminolevulinate Synthetase/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Female , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1/metabolism , Transcriptional Activation
12.
Mol Cell Endocrinol ; 336(1-2): 123-6, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21134412

ABSTRACT

Transformants of mesenchymal stem cells (MSCs) stably expressing steroidogenic factor-1 (SF-1) undergo differentiation into steroidogenic cell-lineages by stimulation with cyclic-adenosine mono-phosphate (cAMP). Another member of NR5A nuclear orphan receptors, Liver-specific receptor homologue-1 (LRH-1), was also able to differentiate MSCs. On the other hand, we found that embryonic stem (ES) cells were hardly induced to differentiate into steroidogenic cell-lineage by the similar treatment. In this study, we developed a novel method to differentiate ES cells into steroidogenic cells. We introduced SF-1 into mouse ES cells at ROSA26 locus under regulation of Tetracycline-off (Tet-off) in order to express SF-1 in the cells at desired period. When SF-1 was induced to express after the ES cells had been differentiated into mesenchymal cell-lineage, steroid hormones were produced from the SF-1 expressing cells. This provides a safer method for supplying sufficient amount of differentiated cells toward future regenerative medicine.


Subject(s)
Cell Differentiation , Cell Lineage , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/cytology , Steroids/biosynthesis , Animals , Humans
13.
Mol Cell Endocrinol ; 336(1-2): 127-32, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21129436

ABSTRACT

Previously, we have demonstrated that mesenchymal stem cells could be differentiated into steroidogenic cells through steroidogenic factor-1 and 8bromo-cAMP treatment. Use of liver receptor homolog-1, another of the nuclear receptor 5A family nuclear receptors, with 8bromo-cAMP also resulted in the differentiation of human mesenchymal stem cells into steroid hormone-producing cells. The same approaches could not be applied to other undifferentiated cells such as embryonic stem cells or embryonal carcinoma cells, because the over-expression of the nuclear receptor 5A family is cytotoxic to these cells. We established embryonic stem cells carrying tetracycline-regulated steroidogenic factor-1 gene at the ROSA26 locus. The embryonic stem cells were first differentiated into a mesenchymal cell lineage by culturing on collagen IV-coated dishes and treating with pulse exposures of retinoic acid before expression of steroidogenic factor-1. Although the untreated embryonic stem cells could not be converted into steroidogenic cells by expression of steroidogenic factor-1 in the absence of leukemia inhibitory factor due to inability of the cells to survive, the differentiated cells could be successfully converted into steroidogenic cells when expression of steroidogenic factor-1 was induced. They exhibited characteristics of adrenocortical-like cells and produced a large amount of corticosterone. These results indicated that pluripotent stem cells could be differentiated into steroidogenic cells by the nuclear receptor 5A family of protein via the mesenchymal cell lineage. This approach may provide a source of cells for future gene therapy for diseases caused by steroidogenesis deficiencies.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Receptors, Cytoplasmic and Nuclear/metabolism , Steroidogenic Factor 1/metabolism , Steroids/biosynthesis , Adrenal Cortex/cytology , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Promoter Regions, Genetic/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Steroidogenic Factor 1/genetics , Tetracycline/pharmacology
14.
Genes Genet Syst ; 86(5): 305-14, 2011.
Article in English | MEDLINE | ID: mdl-22362029

ABSTRACT

Silenced chromatin domains are restricted to specific regions. Eukaryotic chromosomes are organized into discrete domains delimited by domain boundaries. From approximately 6,000 genes in Saccharomyces cerevisiae, we previously isolated 55 boundary genes. In this study, we focus on the molecular function of one of boundary genes, YCR076C/FUB1 (function of boundary), whose function has not been clearly defined in vivo. Biochemical analysis of Fub1p revealed that it interacted with multiple subunits of the 20S proteasome core particle (20S CP). To further clarify the functional link between Fub1p and proteasome, several proteasome mutants were analyzed. Although only 20S CP subunits were isolated as Fub1p interactors, a genetic interaction was also observed for component of 19S regulatory particle (19S RP) suggesting involvement of Fub1p with the whole proteasome. We also analyzed the mechanism of boundary establishment by using proteasome composition factor-deficient strains. Deletion of pre9 and ump1, whose products have effects on the 20S CP, resulted in a decrease in boundary function. Domain analyses of Fub1p identified a minimum functional domain in the C terminus that was essential for boundary establishment and showed a limited sequence homology to the human PSMF1, which is known to inhibit proteasome activity. Finally, boundary assay showed that human PSMF1 also exhibited boundary establishment activity in yeast. Our results defined the functional correlation between Fub1p and PSMF1.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Silencing/physiology , Models, Genetic , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Acetylation , Histones/metabolism , Mass Spectrometry , Proteasome Endopeptidase Complex/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL