Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Mol Cell Endocrinol ; 559: 111808, 2023 01 01.
Article En | MEDLINE | ID: mdl-36309205

Steroidogenesis in ovarian granulosa cells is regulated by the follicle-stimulating hormone (FSH) via transcriptional regulation of its related genes. We herein showed the involvement of the Hippo pathway in this regulation. In KGN granulosa cell, repression of YAP/TAZ activity induced the expression of CYP11A1, HSD3B2, and CYP19A1 in a TEAD-dependent manner without cAMP stimulation. A selective inhibitor of p38 MAP kinase, suppressed YAP/TAZ knockdown-indued the expression of these genes, suggesting this signal could be involved. The expression of these genes was induced by 8Br-cAMP, whereas that of CYR61 and ADATS1, typical YAP/TAZ-TEAD target genes, was suppressed, suggesting that the cellular signaling of cAMP reduced YAP/TAZ-TEAD activity. The constitutively active mutant YAP canceled the FSH- and 8Br-cAMP-mediated induction of these genes in primary rat granulosa and KGN cells, respectively. Moreover, regulation of steroidogenesis-related genes by YAP/TAZ-TEAD was independent of steroidogenic factor 1, a master gene regulator of steroidogenesis. These results suggest that YAP/TAZ-TEAD is a negative regulator of steroidogenesis and that suppression of YAP/TAZ-TEAD activity by FSH is involved in ovarian steroidogenesis.


Transcription Factors , YAP-Signaling Proteins , Female , Rats , Animals , Transcription Factors/genetics , Transcription Factors/metabolism , Granulosa Cells/metabolism , Gene Expression Regulation , Follicle Stimulating Hormone/metabolism
2.
Biosci Biotechnol Biochem ; 86(10): 1438-1447, 2022 Sep 23.
Article En | MEDLINE | ID: mdl-35876648

Histamine (HIST) and other biogenic amines found in fish and fishery products accumulated by the action of bacterial amino acid decarboxylase cannot be decomposed and eliminated by heating or other chemical methods. A simple method for HIST elimination is proposed by a coupling reaction of the fungal amine oxidase (FAO) and bacterial aldehyde oxidase (ALOX) of acetic acid bacteria. As a model reaction, FAO oxidized benzylamine to benzaldehyde, which in turn was oxidized spontaneously to benzoic acid with ALOX. Likely, in HIST elimination, FAO coupled well with ALOX to produce imidazole 4-acetic acid from HIST with an apparent yield of 100%. Imidazole 4-acetaldehyde was not detected in the reaction mixture. In the absence of ALOX, the coupling reaction was incomplete given a number of unidentified substances in the reaction mixture. The proposed coupling enzymatic method may be highly effective to eliminate toxic amines from fish and fishery products.


Carboxy-Lyases , Histamine , Aldehyde Oxidase , Amino Acids , Animals , Bacteria/metabolism , Benzaldehydes , Benzoic Acid , Benzylamines , Biogenic Amines/metabolism , Fishes , Histamine/metabolism
3.
Endocr J ; 67(1): 91-94, 2020 Jan 28.
Article En | MEDLINE | ID: mdl-31534059

Uterine sarcomas are rare and aggressive gynecologic tumors with poor prognosis; therefore, early diagnosis is crucial for therapy. However, it is very difficult to distinguish uterine sarcomas from leiomyomas which are common benign uterine tumors. Therefore, the development of a diagnostic method that utilizes reliable biomarkers to distinguish uterine sarcomas from leiomyomas is important so as to identify the rare tumors. The candidate factors as novel biomarkers were searched for in public databases and a pilot study was performed for confirmation. Growth differentiation factor-15 (GDF15), progranulin, and osteopontin were identified as candidate biomarkers for diagnosing uterine sarcoma. Thus, developing a rapid and easy method to measure these factors could help establish a screening system for uterine sarcomas. In this study, we developed a novel measurement system for these factors using a compact chemical luminescence immunological automatic analyzer POCubeTM. This assay system, which is based on the flow-through membrane immunoassay, completes the whole process and generates results within 15 min. Serum concentrations of these factors measured via POCubeTM correlated well with those measured using enzyme-linked immunosorbent assay (r = 0.994 for GDF15, r = 0.992 for progranulin, and r = 0.976 for osteopontin). The POCubeTM system provides rapid and easy measurement of these factors, thereby facilitating uterine sarcoma diagnosis.


Growth Differentiation Factor 15/blood , Leiomyoma/blood , Osteopontin/blood , Progranulins/blood , Sarcoma/blood , Uterine Neoplasms/blood , Diagnosis, Differential , Female , Humans , Immunoassay , Leiomyoma/diagnosis , Pilot Projects , ROC Curve , Sarcoma/diagnosis , Sensitivity and Specificity , Time Factors , Uterine Neoplasms/diagnosis
4.
Biochem Pharmacol ; 154: 136-147, 2018 08.
Article En | MEDLINE | ID: mdl-29674000

Hypertension is considered as one of the cancer progressive factors, and often found comorbidity in cancer patients. Renin-angiotensin system (RAS) plays an important role in the regulation of blood pressure, and angiotensin II (Ang II) is well known pressor peptide associated with RAS. Ang II has been reported to accelerate progression and metastasis of cancer cells. However, its precise mechanisms have not been fully understood. In this study, we sought to elucidate the mechanisms by which Ang II exacerbates hematogenous metastasis in mouse melanoma cells, focusing the adhesion pathway in vascular endothelial cells. For this purpose, B16/F10 mouse melanoma cells, which do not express the Ang II type 1 receptor (AT1R), were intravenously injected into C57BL/6 mice. Two weeks after cell injection, the number of lung metastatic colonies was significantly higher in the Ang II-treated group (1 µg/kg/min) than in the vehicle-treated group. The AT1R blocker valsartan (40 mg/kg/day), but not the calcium channel blocker amlodipine (5 or 10 mg/kg/day), significantly suppressed the effect of Ang II. In endothelium-specific Agtr1a knockout mice, Ang II-mediated acceleration of lung metastases of melanoma cells was significantly diminished. Ang II treatment significantly increased E-selectin mRNA expression in vascular endothelial cells collected from lung tissues, and thus promoted adherence of melanoma cells to the vascular endothelium. Ang II-accelerated lung metastases of melanoma cells were also suppressed by treatment with anti-E-selectin antibody (20 mg/kg). Taken together, Ang II-treatment exacerbates hematogenous cancer metastasis by promoting E-selectin-mediated adhesion of cancer cells to vascular endothelial cells.


Angiotensin II/toxicity , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/pathology , Lung Neoplasms/pathology , Male , Melanoma, Experimental/chemically induced , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation
5.
Cancer Lett ; 366(2): 182-90, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26164209

Uterine sarcomas are rare and aggressive gynecologic tumors with a poor prognosis because of recurrence and metastasis. However, the mechanisms of uterine sarcoma metastasis are largely unknown. To investigate this mechanism, we developed a novel uterine sarcoma tissue-derived orthotopic and metastatic model in KSN nude mice using a green fluorescent protein stably expressed uterine sarcoma cell line, MES-SA. Histological analysis showed that all orthotopic primary tumors were undifferentiated sarcoma. Primary tumors were characterized by high (18)F-fluorodeoxyglucose uptake with a positive correlation to the number of pulmonary metastases. In addition, we generated uterine sarcoma cell sublines with high or low metastatic potentials by serial in vivo selection. Microarray analysis between orthotopic tumors with high and low metastatic potentials revealed differential expression of genes related to cell proliferation and migration (TNNT1, COL1A2, and ZIC1). Our model would be useful to compensate for the limited clinical cases of uterine sarcoma and to investigate the molecular mechanisms of metastatic uterine sarcoma.


Disease Models, Animal , Lung Neoplasms/secondary , Sarcoma/genetics , Sarcoma/secondary , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Glucose Transport Proteins, Facilitative/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/pharmacokinetics , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Positron-Emission Tomography , Sarcoma/metabolism , Sarcoma/pathology , Transcription Factors/metabolism , Troponin T/metabolism , Tumor Cells, Cultured , Uterine Neoplasms/metabolism
6.
Endocr J ; 62(9): 757-63, 2015.
Article En | MEDLINE | ID: mdl-26135521

Steroid hormones are synthesized from cholesterol in various tissues, mainly in the adrenal glands and gonads. Because these lipid-soluble steroid hormones immediately diffuse through the cells in which they are produced, their secretion directly reflects the activity of the genes related to their production. Progesterone is important not only for luteinization and maintenance of pregnancy, but also as a substrate for most other steroids. Steroidogenic acute regulatory protein (STAR), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), and 3ß-hydroxysteroid dehydrogenase/Δ(5)-Δ(4) isomerase (3ß-HSD) are well-known proteins essential for progesterone production. In addition to them, glutathione S-transferase A1-1 and A3-3 are shown to exert Δ(5)-Δ(4) isomerization activity to produce progesterone in a cooperative fashion with 3ß-HSD. 5-Aminolevulinic acid synthase 1, ferredoxin 1, and ferredoxin reductase also play a role in steroidogenesis as accessory factors. Members of the nuclear receptor 5A (NR5A) family (steroidogenic factor 1 and liver receptor homolog 1) play a crucial role in the transcriptional regulation of these genes. The NR5A family activates these genes by binding to NR5A responsive elements present within their promoter regions, as well as to the elements far from their promoters. In addition, various NR5A-interacting proteins including peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), nuclear receptor subfamily 0, group B, member 1 (DAX-1), and CCAAT/enhancer-binding proteins (C/EBP) are involved in the transcription of NR5A target genes and regulate the transcription either positively or negatively under both basal and tropic hormone-stimulated conditions. In this review, we describe the transcriptional regulation of genes related to progesterone production.


Gene Expression Regulation , Progesterone/biosynthesis , Transcription, Genetic , 17-Hydroxysteroid Dehydrogenases/genetics , Cholesterol Side-Chain Cleavage Enzyme/genetics , Glutathione Transferase/genetics , Humans , Phosphoproteins/genetics , Promoter Regions, Genetic , Steroidogenic Factor 1/genetics
7.
Mol Cell Endocrinol ; 408: 133-7, 2015 Jun 15.
Article En | MEDLINE | ID: mdl-25463758

Steroidogenic factor 1 (SF-1) is a master regulator of adrenal and reproductive development and function. Although SF-1 was identified as a transcriptional regulator for steroid metabolic enzymes, it has been shown that SF-1 also regulates other genes that are involved in various cellular processes. Previously, we showed that introduction of SF-1 into mesenchymal stem cells resulted in the differentiation of these cells to the steroidogenic lineage. By using this method of differentiation, we performed comprehensive analyses to identify the novel SF-1-target genes and components of the SF-1 nuclear complex. Genome-wide analyses with promoter tiling array and DNA microarray identified 10 genes as novel SF-1-target genes including glutathione S-transferase A family, 5-aminolevulinic acid synthase 1 and ferredoxin reductase. Using SF-1 immuno-affinity chromatography of nuclear proteins followed by MS/MS analysis, we identified 24 proteins including CCAAT/enhancer-binding protein ß as components of SF-1 nuclear complex. In this review, we will describe novel roles of the newly identified genes for steroidogenesis.


Cell Nucleus/metabolism , Genetic Association Studies , Multiprotein Complexes/metabolism , Steroidogenic Factor 1/metabolism , Animals , Humans , Models, Biological
8.
Biochim Biophys Acta ; 1839(5): 406-14, 2014 May.
Article En | MEDLINE | ID: mdl-24705138

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.


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
9.
Biochem J ; 460(3): 459-71, 2014 Jun 15.
Article En | MEDLINE | ID: mdl-24635384

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.


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
10.
Biochim Biophys Acta ; 1839(1): 33-42, 2014 Jan.
Article En | MEDLINE | ID: mdl-24321386

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.


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
11.
FASEB J ; 27(8): 3198-208, 2013 Aug.
Article En | MEDLINE | ID: mdl-23650189

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.


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
12.
Endocrinology ; 154(4): 1648-60, 2013 Apr.
Article En | MEDLINE | ID: mdl-23471216

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.


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
13.
Mol Cell Endocrinol ; 370(1-2): 1-10, 2013 May 06.
Article En | MEDLINE | ID: mdl-23435367

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.


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
14.
Mol Cell Endocrinol ; 369(1-2): 42-51, 2013 Apr 30.
Article En | MEDLINE | ID: mdl-23415714

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.


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
15.
Endocrinology ; 153(11): 5522-34, 2012 Nov.
Article En | MEDLINE | ID: mdl-23024262

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.


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
16.
Mar Biotechnol (NY) ; 14(1): 106-19, 2012 Feb.
Article En | MEDLINE | ID: mdl-21748344

The Pacific oyster Crassostrea gigas inhabits the intertidal zone and shows tolerance to stress conditions such as hypoxia and heat shock. Although some information is available about the genes expressed in response to hypoxia, little is known about the molecular mechanism of the regulation of their expression in mollusks, including the Pacific oyster. Hypoxia-inducible factor 1α (HIF-1α) is a master regulator of hypoxia-responsive transcription. In this study, we cloned HIF-α from the oyster and investigated its response to unique stress conditions, including air exposure, for the first time in mollusks. The cDNA of oyster Hif-α is 3,182 bp long, of which 2,094 bp encodes a protein of 698 amino acid residues. Northern and Western blot analysis showed that expression of oyster HIF-α mRNA and protein were induced by air exposure, and that expression was induced periodically during air exposure. In addition, induction of Hif-α mRNA increased by a maximum 8.0-fold by heat shock. Under heat shock at 35°C (lethal temperature for the oyster), however, it was induced later than at 30°C. After recovery from hypoxia and/or heat shock, Hif-α mRNA also upregulated. These data suggest that the oyster has a strategy to induce Hif-α mRNA in order to survive hypoxia and heat shock, and that HIF signaling is necessary for recovery from stress.


Crassostrea/physiology , Gene Expression Regulation/physiology , Hot Temperature , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Air , Amino Acid Sequence , Animals , Antibodies , Base Sequence , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Molecular Sequence Data , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Stress, Physiological/physiology
17.
J Exp Zool A Ecol Genet Physiol ; 315(7): 394-407, 2011 Aug 01.
Article En | MEDLINE | ID: mdl-21455948

The Pacific oyster Crassostrea gigas inhabits the intertidal zone and shows tolerance to various stress conditions such as hypoxia and heat shock. However, little is known about the cellular mechanism of responses to these stresses. Heat shock transcription factor 1 (HSF1) regulates the transcription of several genes, including heat shock proteins (HSPs). In this study, we cloned HSF1 from the oyster and investigated its response to air-exposure. The cDNA of oyster Hsf1 contains 2,931 bp, of which 1,389 bp encode a protein of 463 amino acid residues. Moreover, we found that the oyster has seven novel alternatively spliced isoforms, Hsf1b-h, consisting of insertion A with 48 bp, insertion B with 42 bp and/or insertion C with 42 bp. We determined the sequences of oyster genomic DNA containing Hsf1 insertions A, B and C. The results indicated that eight isoforms of Hsf1 are generated from a single Hsf1 gene by alternative splicing without frameshifting. Real-time PCR analysis showed that Hsf1a is expressed constitutively, and the expression of Hsf1b-h and Hsp70 mRNA is induced by air exposure and/or hypoxia. In addition, we found that 11 putative hypoxia response elements, which are hypoxia-inducible factor 1 (HIF-1) binding sequences, are located in the Hsf1 promoter region. These data suggest that the oyster has an HIF-HSF pathway in which HSPs are induced in an HIF-dependent manner, and that it also has a novel mechanism of alternative splicing of Hsf1 in response to hypoxia.


Alternative Splicing , Crassostrea/metabolism , DNA-Binding Proteins/biosynthesis , Hypoxia/metabolism , Transcription Factors/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Crassostrea/genetics , DNA, Complementary/analysis , DNA-Binding Proteins/genetics , Gene Expression , Heat Shock Transcription Factors , Hypoxia/physiopathology , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics
18.
Mol Cell Endocrinol ; 336(1-2): 123-6, 2011 Apr 10.
Article En | MEDLINE | ID: mdl-21134412

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.


Cell Differentiation , Cell Lineage , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/cytology , Steroids/biosynthesis , Animals , Humans
19.
Mol Cell Endocrinol ; 336(1-2): 127-32, 2011 Apr 10.
Article En | MEDLINE | ID: mdl-21129436

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.


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
20.
Mar Genomics ; 3(1): 19-27, 2010 Mar.
Article En | MEDLINE | ID: mdl-21798193

Calnexin (CNX) and calreticulin (CRT) are endoplasmic reticulum (ER) chaperones. CNX is a type I transmembrane protein and CRT is a soluble CNX homologue. In the ER, CNX and CRT are important for Ca(2+) homeostasis and protein maturation. Here, we describe the full-length cDNA of the first mollusk CNX (cgCNX) and a second mollusk CRT (cgCRT) from the oyster Crassostrea gigas. CgCNX, containing 3255bp, was composed of a 1764bp open reading frame (ORF) that encodes a 588-amino acid protein. CgCRT, containing 1727bp, was composed of a 1242bp ORF that encodes a 414-amino acid protein. CgCNX and cgCRT contains an N-terminal 21- and 16-amino acid sequence, respectively, which is characteristic of a signal sequence. At the C-terminus, cgCRT also contains the KDEL (-Lys-Asp-Glu-Leu) peptide motif suggesting that cgCRT localizes in the ER. Northern blot analysis showed that both cgCNX and cgCRT mRNAs are induced by air exposure. The expression patterns of cgCNX mRNA differed from those of cgCRT during air exposure. This suggests that these two molecular chaperones have different roles in the response to air exposure.


Air , Calnexin/genetics , Calreticulin/genetics , Crassostrea/genetics , Transcriptional Activation , Amino Acid Sequence , Analysis of Variance , Animals , Base Sequence , Blotting, Northern , Calnexin/metabolism , Calreticulin/metabolism , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Endoplasmic Reticulum/metabolism , Hydrogen-Ion Concentration , Molecular Sequence Data , Open Reading Frames/genetics , Pacific Ocean , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
...