1
|
A Short Promoter Region Containing Conserved Regulatory Motifs Is Required for Steroidogenic Acute Regulatory Protein ( Star) Gene Expression in the Mouse Testis. Int J Mol Sci 2022; 23:ijms231912009. [PMID: 36233310 PMCID: PMC9569709 DOI: 10.3390/ijms231912009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022] Open
Abstract
In the testis, Leydig cells produce steroid hormones that are needed to masculinize typical genetic males during fetal development and to initiate and maintain spermatogenesis at puberty and adulthood, respectively. Steroidogenesis is initiated by the transfer of cholesterol from the outer to the inner mitochondrial membrane through the action of steroidogenic acute regulatory protein (STAR). Given its importance for the steroidogenic process, the regulation of STAR gene expression has been the subject of numerous studies. These studies have involved the characterization of key promoter sequences through the identification of relevant transcription factors and the nucleotide motifs (regulatory elements) that they bind. This work has traditionally relied on in vitro studies carried out in cell cultures along with reconstructed promoter sequences. While this approach has been useful for developing models of how a gene might be transcriptionally regulated, one must ultimately validate that these modes of regulation occur in an endogenous context. We have used CRISPR/Cas9 genome editing to modify a short region of the mouse Star promoter (containing a subset of regulatory elements, including conserved CRE, C/EBP, AP1, and GATA motifs) that has been proposed to be critical for Star transcription. Analysis of the resultant mutant mice showed that this short promoter region is indeed required for maximal STAR mRNA and protein levels in the testis. Analysis also showed that both basal and hormone-activated testosterone production in mature mice was unaffected despite significant changes in Star expression. Our results therefore provide the first in vivo validation of regulatory sequences required for Star gene expression.
Collapse
|
2
|
Li Y, Mi P, Wu J, Tang Y, Liu X, Cheng J, Huang Y, Qin W, Cheng CY, Sun F. High Throughput scRNA-Seq Provides Insights Into Leydig Cell Senescence Induced by Experimental Autoimmune Orchitis: A Prominent Role of Interstitial Fibrosis and Complement Activation. Front Immunol 2022; 12:771373. [PMID: 35111154 PMCID: PMC8801941 DOI: 10.3389/fimmu.2021.771373] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 12/28/2022] Open
Abstract
Leydig cells (Lc), located in the interstitial space of the testis between seminiferous tubules, produce 95% of testosterone in male individuals, which is pivotal for male sexual differentiation, spermatogenesis, and maintenance of the male secondary sex characteristics. Lc are prone to senescence in aging testes, resulting in compromised androgen synthesis capability upon aging. However, little is known about whether Lc undergo senescence in a chronic inflammatory environment. To investigate this question, mouse models of experimental autoimmune orchitis (EAO) were used, and Lc were analyzed by high throughput scRNA-Seq. Data were screened and analyzed by correlating signaling pathways with senescence, apoptosis, androgen synthesis, and cytokine/chemokine signaling pathways. EAO did induce Lc senescence, and Lc senescence in turn antagonized androgen synthesis. Based on the correlation screening of pathways inducing Lc senescence, a plethora of pathways were found to play potential roles in triggering Lc senescence during EAO, among which the Arf6 and angiopoietin receptor pathways were highly correlated with senescence signature. Notably, complement and interstitial fibrosis activated by EAO worsened Lc senescence and strongly antagonized androgen synthesis. Furthermore, most proinflammatory cytokines enhanced both senescence and apoptosis in Lc and spermatogonia (Sg) during EAO, and proinflammatory cytokine antagonism of the glutathione metabolism pathway may be key in inducing cellular senescence during EAO.
Collapse
Affiliation(s)
- Yinchuan Li
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, China.,NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Panpan Mi
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, China
| | - Jiabao Wu
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Yunge Tang
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Xiaohua Liu
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Jinmei Cheng
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, China
| | - Yingying Huang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, United States
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, China
| |
Collapse
|
3
|
de Mattos K, Viger RS, Tremblay JJ. Transcription Factors in the Regulation of Leydig Cell Gene Expression and Function. Front Endocrinol (Lausanne) 2022; 13:881309. [PMID: 35464056 PMCID: PMC9022205 DOI: 10.3389/fendo.2022.881309] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/15/2022] [Indexed: 12/28/2022] Open
Abstract
Cell differentiation and acquisition of specialized functions are inherent steps in events that lead to normal tissue development and function. These processes require accurate temporal, tissue, and cell-specific activation or repression of gene transcription. This is achieved by complex interactions between transcription factors that form a unique combinatorial code in each specialized cell type and in response to different physiological signals. Transcription factors typically act by binding to short, nucleotide-specific DNA sequences located in the promoter region of target genes. In males, Leydig cells play a crucial role in sex differentiation, health, and reproductive function from embryonic life to adulthood. To better understand the molecular mechanisms regulating Leydig cell differentiation and function, several transcription factors important to Leydig cells have been identified, including some previously unknown to this specialized cell type. This mini review summarizes the current knowledge on transcription factors in fetal and adult Leydig cells, describing their roles and mechanisms of action.
Collapse
Affiliation(s)
- Karine de Mattos
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, Canada
| | - Robert S. Viger
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Jacques J. Tremblay
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, Canada
- *Correspondence: Jacques J. Tremblay,
| |
Collapse
|
4
|
Smith LIF, Zhao Z, Walker J, Lightman S, Spiga F. Activation and expression of endogenous CREB-regulated transcription coactivators (CRTC) 1, 2 and 3 in the rat adrenal gland. J Neuroendocrinol 2021; 33:e12920. [PMID: 33314405 PMCID: PMC7900988 DOI: 10.1111/jne.12920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 12/30/2022]
Abstract
The activation and nuclear translocation of cAMP-response element binding protein (CREB)-regulated transcription coactivator (CRTC)2 occurs in the rat adrenal gland, in response to adrenocorticotrophic hormone (ACTH) and stressors, and has been implicated in the transcriptional regulation of steroidogenic acute regulatory protein (StAR). We have recently demonstrated the activation of CRTC isoforms, CRTC1 and CRTC3, in adrenocortical cell lines. In the present study, we aimed to determine the activation and expression of the three CRTC isoforms in vivo in relation to Star transcription, under basal conditions and following a robust endotoxic stress challenge. Rat adrenal glands and blood plasma were collected following i.v. administration of either an ultradian-sized pulse of ACTH or administration of lipopolysaccharide, as well as under unstressed conditions across the 24-hour period. Plasma ACTH and corticosterone (CORT) were measured and the adrenal glands were processed for measurement of protein by western immunoblotting, RNA by a quantitative reverse transcriptase-polymerase chain reaction and association of CRTC2 and CRTC3 with the Star promoter by chromatin immunoprecipitation. An increase in nuclear localisation of CRTC2 and CRTC3 followed increases in both ultradian and endotoxic stress-induced plasma ACTH, and this was associated with increased CREB phosphorylation and corresponding increases in Star transcription. Both CRTC2 and CRTC3 were shown to associate with the Star promoter, with the dynamics of CRTC3 binding corresponding to that of nuclear changes in protein levels. CRTC isoforms show little variation in ultradian expression or variation across 24 hours, although evidence of long-term down-regulation following endotoxic stress was found. We conclude that co-transcription factors CRTC2 and, more clearly, CRTC3 appear to act alongside phosphorylated CREB in the generation of ultradian pulses of Star transcription, essential for the maintenance of basal StAR expression. Similarly, our findings suggest CRTC2 and CRTC3 mediate Star transcriptional initiation following an endotoxic stressor; however, other transcription factors are likely to be responsible for the long-term up-regulation of adrenal Star transcription.
Collapse
Affiliation(s)
- Lorna I. F. Smith
- Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| | - Zidong Zhao
- Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| | - Jamie Walker
- Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
- College of Engineering, Mathematics and Physical SciencesUniversity of ExeterExeterUK
- EPSRC Centre for Predictive Modelling in HealthcareUniversity of ExeterExeterUK
| | - Stafford Lightman
- Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| | - Francesca Spiga
- Bristol Medical School: Translational Health SciencesUniversity of BristolBristolUK
| |
Collapse
|
5
|
Selvaraj V, Stocco DM, Clark BJ. Current knowledge on the acute regulation of steroidogenesis. Biol Reprod 2018; 99:13-26. [PMID: 29718098 PMCID: PMC6044331 DOI: 10.1093/biolre/ioy102] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/23/2018] [Accepted: 04/26/2018] [Indexed: 12/31/2022] Open
Abstract
How rapid induction of steroid hormone biosynthesis occurs in response to trophic hormone stimulation of steroidogenic cells has been a subject of intensive investigation for approximately six decades. A key observation made very early was that acute regulation of steroid biosynthesis required swift and timely synthesis of a new protein whose role appeared to be involved in the delivery of the substrate for all steroid hormones, cholesterol, from the outer to the inner mitochondrial membrane where the process of steroidogenesis begins. It was quickly learned that this transfer of cholesterol to the inner mitochondrial membrane was the regulated and rate-limiting step in steroidogenesis. Following this observation, the quest for this putative regulator protein(s) began in earnest in the late 1950s. This review provides a history of this quest, the candidate proteins that arose over the years and facts surrounding their rise or decline. Only two have persisted-translocator protein (TSPO) and the steroidogenic acute regulatory protein (StAR). We present a detailed summary of the work that has been published for each of these two proteins, the specific data that has appeared in support of their role in cholesterol transport and steroidogenesis, and the ensuing observations that have arisen in recent years that have refuted the role of TSPO in this process. We believe that the only viable candidate that has been shown to be indispensable is the StAR protein. Lastly, we provide our view on what may be the most important questions concerning the acute regulation of steroidogenesis that need to be asked in future.
Collapse
Affiliation(s)
- Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, USA
| | - Douglas M Stocco
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Barbara J Clark
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
6
|
Yang Y, Luo J, Yu D, Zhang T, Lin Q, Li Q, Wu X, Su Z, Zhang Q, Xiang Q, Huang Y. Vitamin A Promotes Leydig Cell Differentiation via Alcohol Dehydrogenase 1. Front Endocrinol (Lausanne) 2018; 9:644. [PMID: 30420837 PMCID: PMC6216111 DOI: 10.3389/fendo.2018.00644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/10/2018] [Indexed: 12/02/2022] Open
Abstract
Vitamin A (retinol) is important for multiple functions in mammals. In testis, the role of vitamin A in the regulation of testicular functions is clearly involved in rodents. It is essential for sperm production. Vitamin A deficiency adversely affects testosterone secretion. Adult Leydig cells are responsible for testosterone production in male. The role of vitamin A in regulating the differentiation of Leydig cells is still unknown. In this study, we explored the roles and underlying mechanisms of vitamin A in Leydig cell differentiation. We found that vitamin A could regulate the Leydig cells differentiation. Leydig cell differentiation is adversely affected in mice maintained on a vitamin A-free diet. This effect is mediated by alcohol dehydrogenase 1 (ADH1). ADH1 could increase retinoic acid (RA) synthesis, then RA facilitates Leydig cell differentiation by activating the steroidogenic factor 1 gene (Nr5a1) promoter activity, which consequently promotes Leydig cell specific gene expression, resulting in progenitor Leydig cells differentiation into functional Leydig cells. This is the first study connecting a metabolic enzyme of retinol (ADH1) to the the regulation of Leydig cell differentiation, which will provide experimental evidence for the development of therapeutics to promote Leydig regeneration through the administration of a RA signaling regulator or a vitamin A supplement.
Collapse
Affiliation(s)
- Yan Yang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
- Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Jiao Luo
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Dan Yu
- Department of Pharmacology, Jinan University, Guangzhou, China
| | - Tiantian Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qilian Lin
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Quan Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Xupeng Wu
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Zhijian Su
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qihao Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qi Xiang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
- Department of Pharmacology, Jinan University, Guangzhou, China
| | - Yadong Huang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology, Jinan University, Guangzhou, China
- Department of Pharmacology, Jinan University, Guangzhou, China
- *Correspondence: Yadong Huang
| |
Collapse
|
7
|
Lee J, Jefcoate C. Monitoring of Dual CRISPR/Cas9-Mediated Steroidogenic Acute Regulatory Protein Gene Deletion and Cholesterol Accumulation Using High-Resolution Fluorescence In Situ Hybridization in a Single Cell. Front Endocrinol (Lausanne) 2017; 8:289. [PMID: 29118738 PMCID: PMC5660980 DOI: 10.3389/fendo.2017.00289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 10/11/2017] [Indexed: 02/02/2023] Open
Abstract
Recent advances in fluorescence microscopy, coupled with CRISPR/Cas9 gene editing technology, provide opportunities for understanding gene regulation at the single-cell level. The application of direct imaging shown here provides an in situ side-by-side comparison of CRISPR/Cas9-edited cells and adjacent unedited cells. We apply this methodology to the steroidogenic acute regulatory protein (StAR) gene in Y-1 adrenal cells and MA-10 testis cells. StAR is a gatekeeper protein that controls the access of cholesterol from the cytoplasm to the inner mitochondria. The loss of this mitochondrial cholesterol transfer mediator rapidly increases lipid droplets in cells, as seen in StAR-/- mice. Here, we describe a dual CRISPR/Cas9 strategy marked by GFP/mCherry expression that deletes StAR activity within 12 h. We used single-molecule fluorescence in situ hybridization (sm-FISH) imaging to directly monitor the time course of gene editing in single cells. We achieved StAR gene deletion at high efficiency dual gRNA targeting to the proximal promoter and exon 2. Seventy percent of transfected cells showed a slow DNA deletion as measured by PCR, and loss of Br-cAMP stimulated transcription. This DNA deletion was seen by sm-FISH in both loci of individual cells relative to non-target Cyp11a1 and StAR exon 7. sm-FISH also distinguishes two effects on stimulated StAR expression without this deletion. Br-cAMP stimulation of primary and spliced StAR RNA at the gene loci were removed within 4 h in this dual CRISPR/Cas9 strategy before any effect on cytoplasmic mRNA and protein occurred. StAR mRNA disappeared between 12 and 24 h in parallel with this deletion, while cholesterol ester droplets increased fourfold. These alternative changes match distinct StAR expression processes. This dual gRNA and sm-FISH approach to CRISPR/Cas9 editing facilitates rapid testing of editing strategies and immediate assessment of single-cell adaptation responses without the perturbation of clonal expansion procedures.
Collapse
Affiliation(s)
- Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, United States
- *Correspondence: Jinwoo Lee, ; Colin Jefcoate,
| | - Colin Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI, United States
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI, United States
- Molecular and Cellular Pharmacology, University of Wisconsin, Madison, WI, United States
- *Correspondence: Jinwoo Lee, ; Colin Jefcoate,
| |
Collapse
|
8
|
Lee J, Foong YH, Musaitif I, Tong T, Jefcoate C. Analysis of specific RNA in cultured cells through quantitative integration of q-PCR and N-SIM single cell FISH images: Application to hormonal stimulation of StAR transcription. Mol Cell Endocrinol 2016; 429:93-105. [PMID: 27091298 PMCID: PMC6181224 DOI: 10.1016/j.mce.2016.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/02/2016] [Accepted: 04/02/2016] [Indexed: 12/21/2022]
Abstract
The steroidogenic acute regulatory protein (StAR) has been proposed to serve as the switch that can turn on/off steroidogenesis. We investigated the events that facilitate dynamic StAR transcription in response to cAMP stimulation in MA-10 Leydig cells, focusing on splicing anomalies at StAR gene loci. We used 3' reverse primers in a single reaction to respectively quantify StAR primary (p-RNA), spliced (sp-RNA/mRNA), and extended 3' untranslated region (UTR) transcripts, which were quantitatively imaged by high-resolution fluorescence in situ hybridization (FISH). This approach delivers spatio-temporal resolution of initiation and splicing at single StAR loci, and transfers individual mRNA molecules to cytoplasmic sites. Gene expression was biphasic, initially showing slow splicing, transitioning to concerted splicing. The alternative 3.5-kb mRNAs were distinguished through the use of extended 3'UTR probes, which exhibited distinctive mitochondrial distribution. Combining quantitative PCR and FISH enables imaging of localization of RNA expression and analysis of RNA processing rates.
Collapse
Affiliation(s)
- Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States.
| | - Yee Hoon Foong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Ibrahim Musaitif
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Tiegang Tong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Colin Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States
| |
Collapse
|
9
|
Ruggiero C, Lalli E. Impact of ACTH Signaling on Transcriptional Regulation of Steroidogenic Genes. Front Endocrinol (Lausanne) 2016; 7:24. [PMID: 27065945 PMCID: PMC4810002 DOI: 10.3389/fendo.2016.00024] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 01/12/2023] Open
Abstract
The trophic peptide hormone adrenocorticotropic (ACTH) stimulates steroid hormone biosynthesis evoking both a rapid, acute response and a long-term, chronic response, via the activation of cAMP/protein kinase A (PKA) signaling. The acute response is initiated by the mobilization of cholesterol from lipid stores and its delivery to the inner mitochondrial membrane, a process that is mediated by the steroidogenic acute regulatory protein. The chronic response results in the increased coordinated transcription of genes encoding steroidogenic enzymes. ACTH binding to its cognate receptor, melanocortin 2 receptor (MC2R), stimulates adenylyl cyclase, thus inducing cAMP production, PKA activation, and phosphorylation of specific nuclear factors, which bind to target promoters and facilitate coactivator protein recruitment to direct steroidogenic gene transcription. This review provides a general view of the transcriptional control exerted by the ACTH/cAMP system on the expression of genes encoding for steroidogenic enzymes in the adrenal cortex. Special emphasis will be given to the transcription factors required to mediate ACTH-dependent transcription of steroidogenic genes.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Valbonne, France
- Laboratoire International Associé (LIA) CNRS NEOGENEX, Valbonne, France
- Université de Nice, Valbonne, France
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, Valbonne, France
- Laboratoire International Associé (LIA) CNRS NEOGENEX, Valbonne, France
- Université de Nice, Valbonne, France
| |
Collapse
|
10
|
Vasquez YM, Wu SP, Anderson ML, Hawkins SM, Creighton CJ, Ray M, Tsai SY, Tsai MJ, Lydon JP, DeMayo FJ. Endometrial Expression of Steroidogenic Factor 1 Promotes Cystic Glandular Morphogenesis. Mol Endocrinol 2016; 30:518-32. [PMID: 27018534 DOI: 10.1210/me.2015-1215] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Epigenetic silencing of steroidogenic factor 1 (SF1) is lost in endometriosis, potentially contributing to de novo local steroidogenesis favoring inflammation and growth of ectopic endometrial tissue. In this study, we examine the impact of SF1 expression in the eutopic uterus by a novel mouse model that conditionally expresses SF1 in endometrium. In vivo SF1 expression promoted the development of enlarged endometrial glands and attenuated estrogen and progesterone responsiveness. Endometriosis induction by autotransplantation of uterine tissue to the mesenteric membrane resulted in the increase in size of ectopic lesions from SF1-expressing mice. By integrating the SF1-dependent transcriptome with the whole genome binding profile of SF1, we identified uterine-specific SF1-regulated genes involved in Wingless and Progesterone receptor-Hedgehog-Chicken ovalbumin upstream promoter transcription factor II signaling for gland development and epithelium-stroma interaction, respectively. The present results indicate that SF1 directly contributes to the abnormal uterine gland morphogenesis, an inhibition of steroid hormone signaling and activation of an immune response, in addition to previously postulated estrogen production.
Collapse
Affiliation(s)
- Yasmin M Vasquez
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - San-Pin Wu
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Matthew L Anderson
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Shannon M Hawkins
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Chad J Creighton
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Madhumita Ray
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - John P Lydon
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Francesco J DeMayo
- Department of Molecular and Cellular Biology (Y.M.V., S.Y.T., M.-J.T., J.P.L., F.J.D.), Baylor College of Medicine, Houston, Texas 77030; Department of Obstetrics and Gynecology (M.L.A., S.M.H.), Baylor College of Medicine, Houston, Texas 77030; Dan L. Duncan Cancer Center (M.L.A., C.J.C.), Division of Biostatistics, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030; and Pregnancy and Female Reproduction Group (S.-P.W., M.R., M.J.D.), National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| |
Collapse
|
11
|
Yang Y, Su Z, Xu W, Luo J, Liang R, Xiang Q, Zhang Q, Ge RS, Huang Y. Directed mouse embryonic stem cells into leydig-like cells rescue testosterone-deficient male rats in vivo. Stem Cells Dev 2014; 24:459-70. [PMID: 25340537 DOI: 10.1089/scd.2014.0370] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The primary function of Leydig cells is to secrete testosterone, which is critical in the regulation of male reproduction and development. Low levels of testosterone will lead to male hypogonadism. Stem cell-derived Leydig cell transplantation may be a promising alternative therapy for male hypogonadism. Thus far, others have reported that Leydig-like cells can be derived from mesenchymal stem cells, embryonic stem cells (ESCs), and induced pluripotent stem cells. However, the efficiency of the differentiating Leydig cells remains low, and progress toward generating functional adult Leydig cells (ALCs) is limited. Herein, we describe a robust method of directing differentiation of mouse embryonic stem cells (mESCs) into Leydig-like cells in vitro by overexpression of the transcription factor steroidogenic factor-1 (SF-1) and treatment with a combination of 8-Bromoadenosine-3',5'-cyclic monophosphate and forskolin. These differentiated cells express mRNA encoding the steroidogenic enzymes and produce progesterone and testosterone. Importantly, when transplanted into male rats that had their original Leydig cells selectively eliminated by ethylene dimethanesulfonate, these in vitro-derived Leydig-like cells further developed into functional ALCs that rescued serum testosterone levels. These data provide evidence that mESCs can be induced to differentiate into Leydig-like cells in vitro, which can develop in the in vivo microenvironment.
Collapse
Affiliation(s)
- Yan Yang
- 1 Department of Cell Biology, College of Life Science and Technology, Jinan University , Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Duarte A, Castillo AF, Podestá EJ, Poderoso C. Mitochondrial fusion and ERK activity regulate steroidogenic acute regulatory protein localization in mitochondria. PLoS One 2014; 9:e100387. [PMID: 24945345 PMCID: PMC4063759 DOI: 10.1371/journal.pone.0100387] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 05/27/2014] [Indexed: 11/21/2022] Open
Abstract
The rate-limiting step in the biosynthesis of steroid hormones, known as the transfer of cholesterol from the outer to the inner mitochondrial membrane, is facilitated by StAR, the Steroidogenic Acute Regulatory protein. We have described that mitochondrial ERK1/2 phosphorylates StAR and that mitochondrial fusion, through the up-regulation of a fusion protein Mitofusin 2, is essential during steroidogenesis. Here, we demonstrate that mitochondrial StAR together with mitochondrial active ERK and PKA are necessary for maximal steroid production. Phosphorylation of StAR by ERK is required for the maintenance of this protein in mitochondria, observed by means of over-expression of a StAR variant lacking the ERK phosphorylation residue. Mitochondrial fusion regulates StAR levels in mitochondria after hormone stimulation. In this study, Mitofusin 2 knockdown and mitochondrial fusion inhibition in MA-10 Leydig cells diminished StAR mRNA levels and concomitantly mitochondrial StAR protein. Together our results unveil the requirement of mitochondrial fusion in the regulation of the localization and mRNA abundance of StAR. We here establish the relevance of mitochondrial phosphorylation events in the correct localization of this key protein to exert its action in specialized cells. These discoveries highlight the importance of mitochondrial fusion and ERK phosphorylation in cholesterol transport by means of directing StAR to the outer mitochondrial membrane to achieve a large number of steroid molecules per unit of StAR.
Collapse
Affiliation(s)
- Alejandra Duarte
- Institute of Biomedical Investigations (INBIOMED, UBA-CONICET), Department of Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Ana Fernanda Castillo
- Institute of Biomedical Investigations (INBIOMED, UBA-CONICET), Department of Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Ernesto J. Podestá
- Institute of Biomedical Investigations (INBIOMED, UBA-CONICET), Department of Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Cecilia Poderoso
- Institute of Biomedical Investigations (INBIOMED, UBA-CONICET), Department of Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
13
|
Urlep Z, Rozman D. The Interplay between Circadian System, Cholesterol Synthesis, and Steroidogenesis Affects Various Aspects of Female Reproduction. Front Endocrinol (Lausanne) 2013; 4:111. [PMID: 24065951 PMCID: PMC3778439 DOI: 10.3389/fendo.2013.00111] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 08/13/2013] [Indexed: 01/22/2023] Open
Abstract
Circadian aspect of reproduction has gained much attention in recent years. In mammals, it is very important that the timing of greatest sexual motivation is in line with the highest fertility. Peripheral clocks have been found to reside also in reproductive organs, such as the uterus and ovary. The timing signal from the suprachiasmatic nucleus is suggested to be transmitted via hormonal and neural mechanisms, and could thus mediate circadian expression of target genes in these organs. In turn, estrogens from the ovary have been found to signal back to the hypothalamus, completing the feedback loop. In this review we will focus on the interplay between clock and estrogens. Estradiol has been directly linked with expression of Per1 and Per2 in the uterus. CLOCK, on the other hand, has been shown to alter estradiol signaling. We also present the idea that cholesterol could play a vital role in the regulation of reproduction. Cholesterol synthesis itself is circadially regulated and has been found to interfere with steroidogenesis in the ovary on the molecular level. This review presents a systems view on how the interplay between circadian clock, steroidogenesis, and cholesterol synthesis affect various aspects of mammalian reproduction.
Collapse
Affiliation(s)
- Ziga Urlep
- Center for Functional Genomics and Bio-Chips, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Institute for Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Damjana Rozman, Center for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia e-mail:
| |
Collapse
|
14
|
Hu Y, Dong C, Chen M, Lu J, Han X, Qiu L, Chen Y, Qin J, Li X, Gu A, Xia Y, Sun H, Li Z, Wang Y. Low-dose monobutyl phthalate stimulates steroidogenesis through steroidogenic acute regulatory protein regulated by SF-1, GATA-4 and C/EBP-beta in mouse Leydig tumor cells. Reprod Biol Endocrinol 2013; 11:72. [PMID: 23889939 PMCID: PMC3734203 DOI: 10.1186/1477-7827-11-72] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 07/16/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The ubiquitous use of dibutyl phthalate (DBP), one of the most widely used plasticizers, results in extensive exposure to humans and the environment. DBP and its major metabolite, monobutyl phthalate (MBP), may alter steroid biosynthesis and their exposure may lead to damage to male reproductive function. Low-doses of DBP/MBP may result in increased steroidogenesis in vitro and in vivo. However, the mechanisms of possible effects of low-dose MBP on steroidogenesis remain unclear. The aim of present study was to elaborate the role of transcription factors and steroidogenic acute regulatory protein in low-dose MBP-induced distruption of steroidogenesis in mouse Leydig tumor cells (MLTC-1 cells). METHODS In the present study, MLTC-1 cells were cultured in RPMI 1640 medium supplemented with 2 g/L sodium bicarbonate. Progesterone level was examined by I125-pregesterone Coat-A-Count radioimmunoassay (RIA) kits. mRNA and protein levels were assessed by reverse transcription-polymerase chain reaction (RT-PCR) and western blot, respectively. DNA-binding of several transcription factors was examined by electrophoretic mobility shift assay (EMSA). RESULTS In this study, various doses of MBP (0, 10(-9), 10(-8), 10(-7), or 10(-6) M) were added to the medium followed by stimulation of MLTC-1 cells with human chorionic gonadotrophin (hCG). The results showed that MBP increased progesterone production and steroidogenic acute regulatory protein (StAR) mRNA and protein levels. However, the protein levels of cytochrome P450scc and 3 beta-hydroxy-steroid dehydrogenase (3 beta-HSD) were unchanged after MBP treatment. EMSA assay showed that DNA-binding of steroidogenic factors 1(SF-1), GATA-4 and CCAAT/enhancer binding protein-beta (C/EBP-beta) was increased in a dose-dependent manner after MBP exposure. Western blot tests were next employed and confirmed that the protein levels of SF-1, GATA-4 and C/EBP-beta were also increased. Additionally, western blot tests confirmed the expression of DAX-1, negative factor of SF-1, was dose-dependently down regulated after MBP exposure, which further confirmed the role of SF-1 in MBP-stimulated steroid biosynthesis. CONCLUSIONS In conclusion, we firstly delineated the regulation of StAR by transcription factors including SF-1, GATA-4 and C/EBP-beta maybe critical mechanism involved in low-dose MBP-stimulated steroidogenesis.
Collapse
Affiliation(s)
- Yanhui Hu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Congcong Dong
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jing Lu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lianglin Qiu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yansu Chen
- Department of Molecular Cell Biology and Toxicology, Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jingjing Qin
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaocheng Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Hong Sun
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 211166, China
| | - Zhong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yubang Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
15
|
Meier RK, Clark BJ. Angiotensin II-dependent transcriptional activation of human steroidogenic acute regulatory protein gene by a 25-kDa cAMP-responsive element modulator protein isoform and Yin Yang 1. Endocrinology 2012; 153:1256-68. [PMID: 22253417 PMCID: PMC3281547 DOI: 10.1210/en.2011-1744] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Transcriptional activation of the steroidogenic acute regulatory protein (STAR) gene is a critical component in the angiotensin II (Ang II)-dependent increase in aldosterone biosynthesis in the adrenal gland. The purpose of this study was to define the molecular mechanisms that mediate the Ang II-dependent increase in STARD1 gene (STAR) expression in H295R human adrenocortical cells. Mutational analysis of the STAR proximal promoter revealed that a nonconsensus cAMP-responsive element located at -78 bp relative to the transcription start site (-78CRE) is required for the Ang II-stimulated STAR reporter gene activity. DNA immunoaffinity chromatography identified a 25-kDa cAMP-responsive element modulator isoform and Yin Yang 1 (YY1) as -78CRE DNA-binding proteins, and Ang II treatment of H295R cells increased expression of that 25-kDa CREM isoform. Small interfering RNA silencing of CREM and YY1 attenuated the Ang II-dependent increases in STAR reporter gene activity and STAR mRNA levels. Conversely, overexpression of CREM and YY1 in COS-1 cells resulted in transactivation of STAR reporter gene activity. Chromatin immunoprecipitation analysis demonstrated recruitment of CREM and YY1 to the STAR promoter along with increased association of the coactivator cAMP response element-binding protein-binding protein (CBP) and increased phosphorylated RNA polymerase II after Ang II treatment. Together our data reveal that the Ang II-stimulated increase in STAR expression in H295R cells requires 25 kDa CREM and YY1. The recruitment of these transcription factors to the STAR proximal promoter results in association of CBP and activation of RNA polymerase II leading to increased STAR transcription.
Collapse
Affiliation(s)
- Renate K Meier
- Department of Biochemistry and Molecular Biology, University of Louisville, School of Medicine, Louisville, Kentucky 40292, USA
| | | |
Collapse
|
16
|
Mizutani T, Yazawa T, Ju Y, Imamichi Y, Uesaka M, Inaoka Y, Matsuura K, Kamiki Y, Oki M, Umezawa A, Miyamoto K. Identification of a novel distal control region upstream of the human steroidogenic acute regulatory protein (StAR) gene that participates in SF-1-dependent chromatin architecture. J Biol Chem 2010; 285:28240-51. [PMID: 20601698 DOI: 10.1074/jbc.m110.129510] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
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.
Collapse
Affiliation(s)
- Tetsuya Mizutani
- Department of Biochemistry, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Barsoum IB, Yao HHC. Fetal Leydig cells: progenitor cell maintenance and differentiation. ACTA ACUST UNITED AC 2009; 31:11-5. [PMID: 19875489 DOI: 10.2164/jandrol.109.008318] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In most eutherian mammals, sexually dimorphic masculinization is established by androgen-producing fetal Leydig cells in the embryonic testis. Fetal Leydig cells, which lack expression of the testis-determining gene SRY, arise after the appearance of SRY-expressing Sertoli cells. Therefore, the appearance and differentiation of fetal Leydig cells are probably regulated by factors derived from Sertoli cells. Results from mouse genetic models have revealed that maintenance and differentiation of fetal Leydig cell population depends upon a balance between differentiation-promoting and differentiation-suppressing mechanisms. Although paracrine signaling via Sertoli cell-derived Hedgehog ligands is necessary and sufficient for fetal Leydig cell formation, cell-cell interaction via Notch signaling and intracellular transcription factors such as POD1 are implicated as suppressors of fetal Leydig cell differentiation. This review provides a model that summarizes the recent findings in fetal Leydig cell development.
Collapse
Affiliation(s)
- Ivraym B Barsoum
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61802, USA
| | | |
Collapse
|
18
|
Barsoum IB, Bingham NC, Parker KL, Jorgensen JS, Yao HHC. Activation of the Hedgehog pathway in the mouse fetal ovary leads to ectopic appearance of fetal Leydig cells and female pseudohermaphroditism. Dev Biol 2009; 329:96-103. [PMID: 19268447 PMCID: PMC2673990 DOI: 10.1016/j.ydbio.2009.02.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 02/18/2009] [Accepted: 02/19/2009] [Indexed: 11/16/2022]
Abstract
Proper cell fate determination in mammalian gonads is critical for the establishment of sexual identity. The Hedgehog (Hh) pathway has been implicated in cell fate decision for various organs, including gonads. Desert Hedgehog (Dhh), one of the three mammalian Hh genes, has been implicated with other genes in the establishment of mouse fetal Leydig cells. To investigate whether Hh alone is sufficient to induce fetal Leydig cell differentiation, we ectopically activated the Hh pathway in Steroidogenic factor 1 (SF1)-positive somatic cell precursors of fetal ovaries. Hh activation transformed SF1-positive somatic ovarian cells into functional fetal Leydig cells. These ectopic fetal Leydig cells produced androgens and insulin-like growth factor 3 (INLS3) that cause virilization of female embryos and ovarian descent. However, the female reproductive system remained intact, indicating a typical example of female pseudohermaphroditism. The appearance of fetal Leydig cells was a direct consequence of Hh activation as evident by the absence of other testicular components in the affected ovary. This study provides not only insights into mechanisms of cell lineage specification in gonads, but also a model to understand defects in sexual differentiation.
Collapse
Affiliation(s)
- Ivraym B. Barsoum
- Department of Cell and Developmental Biology, University of Illinois at Urbana- Champaign, IL 61820
| | - Nathan C. Bingham
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235
| | - Keith L. Parker
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53706
| | - Humphrey H-C Yao
- Department of Veterinary Biosciences, University of Illinois at Urbana-Champaign, IL 61820
| |
Collapse
|
19
|
Manna PR, Dyson MT, Stocco DM. Role of basic leucine zipper proteins in transcriptional regulation of the steroidogenic acute regulatory protein gene. Mol Cell Endocrinol 2009; 302:1-11. [PMID: 19150388 PMCID: PMC5006949 DOI: 10.1016/j.mce.2008.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/15/2008] [Accepted: 12/15/2008] [Indexed: 01/23/2023]
Abstract
The regulation of steroidogenic acute regulatory protein (StAR) gene transcription by cAMP-dependent mechanisms occurs in the absence of a consensus cAMP response element (CRE, TGACGTGA). This regulation is coordinated by multiple transcription factors that bind to sequence-specific elements located approximately 150 bp upstream of the transcription start site. Among the proteins that bind within this region, the basic leucine zipper (bZIP) family of transcription factors, i.e. CRE binding protein (CREB)/CRE modulator (CREM)/activating transcription factor (ATF), activator protein 1 (AP-1; Fos/Jun), and CCAAT enhancer binding protein beta (C/EBPbeta), interact with an overlapping region (-81/-72 bp) in the StAR promoter, mediate stimulus-transcription coupling of cAMP signaling and play integral roles in regulating StAR gene expression. These bZIP proteins are structurally similar and bind to DNA sequences as dimers; however, they exhibit discrete transcriptional activities, interact with several transcription factors and other properties that contribute in their regulatory functions. The 5'-flanking -81/-72 bp region of the StAR gene appears to function as a key element within a complex cAMP response unit by binding to different bZIP members, and the StAR promoter displays variable states of cAMP responsivity contingent upon the occupancy of these cis-elements with these transcription factors. The expression and activities of CREB/CREM/ATF, Fos/Jun and C/EBPbeta have been demonstrated to be mediated by a plethora of extracellular signals, and the phosphorylation of these proteins at several Ser and Thr residues allows recruitment of the transcriptional coactivator CREB binding protein (CBP) or its functional homolog p300 to the StAR promoter. This review will focus on the current level of understanding of the roles of selective bZIP family proteins within the complex series of processes involved in regulating StAR gene transcription.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | |
Collapse
|
20
|
Manna PR, Dyson MT, Stocco DM. Regulation of the steroidogenic acute regulatory protein gene expression: present and future perspectives. Mol Hum Reprod 2009; 15:321-33. [PMID: 19321517 DOI: 10.1093/molehr/gap025] [Citation(s) in RCA: 229] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Steroid hormones are synthesized in the adrenal gland, gonads, placenta and brain and are critical for normal reproductive function and bodily homeostasis. The steroidogenic acute regulatory (StAR) protein regulates the rate-limiting step in steroid biosynthesis, i.e. the delivery of cholesterol from the outer to the inner mitochondrial membrane. The expression of the StAR protein is predominantly regulated by cAMP-dependent mechanisms in the adrenal and gonads. Whereas StAR plays an indispensable role in the regulation of steroid biosynthesis, a complete understanding of the regulation of its expression and function in steroidogenesis is not available. It has become clear that the regulation of StAR gene expression is a complex process that involves the interaction of a diversity of hormones and multiple signaling pathways that coordinate the cooperation and interaction of transcriptional machinery, as well as a number of post-transcriptional mechanisms that govern mRNA and protein expression. However, information is lacking on how the StAR gene is regulated in vivo such that it is expressed at appropriate times during development and is confined to the steroidogenic cells. Thus, it is not surprising that the precise mechanism involved in the regulation of StAR gene has not yet been established, which is the key to understanding the regulation of steroidogenesis in the context of both male and female development and function.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | | |
Collapse
|
21
|
Yivgi-Ohana N, Sher N, Melamed-Book N, Eimerl S, Koler M, Manna PR, Stocco DM, Orly J. Transcription of steroidogenic acute regulatory protein in the rodent ovary and placenta: alternative modes of cyclic adenosine 3', 5'-monophosphate dependent and independent regulation. Endocrinology 2009; 150:977-89. [PMID: 18845640 PMCID: PMC2732291 DOI: 10.1210/en.2008-0541] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Steroid hormone synthesis is a vital function of the adrenal cortex, serves a critical role in gonadal function, and maintains pregnancy if normally executed in the placenta. The substrate for the synthesis of all steroid hormones is cholesterol, and its conversion to the first steroid, pregnenolone, by the cholesterol side-chain cleavage cytochrome P450 (CYP11A1) enzyme complex takes place in the inner mitochondrial membranes. Steroidogenic acute regulatory protein (STAR) facilitates the rate-limiting transfer of cholesterol from the outer mitochondrial membrane to CYP11A1 located in the inner organelle membranes. The current study explored the mechanisms controlling transcription of the Star gene in primary cell cultures of mouse placental trophoblast giant cells and rat ovarian granulosa cells examined throughout the course of their functional differentiation. Our findings show that the cis-elements required for Star transcription in the rodent placenta and the ovary are centered in a relatively small proximal region of the promoter. In placental trophoblast giant cells, cAMP is required for activation of the Star promoter, and the cis-elements mediating a maximal response were defined as cAMP response element 2 and GATA. EMSA studies show that placental cAMP-responsive element binding protein (CREB)-1 and activating transcription factor-2 (ATF2) bind to a -81/-78 sequence, whereas GATA-2 binds to a -66/-61 sequence. In comparison, patterns of Star regulation in the ovary suggested tissue-specific and developmental controlled modes of Star transcription. During the follicular phase, FSH/cAMP induced CREB-1 dependent activity, whereas upon luteinization STAR expression becomes cAMP and CREB independent, a functional shift conferred by FOS-related antigen-2 displacement of CREB-1 binding, and the appearance of a new requirement for CCAAT enhancer-binding protein beta and steroidogenic factor 1 that bind to upstream elements (-117/-95). These findings suggest that during evolution, the promoters of the Star gene acquired nonconsensus sequence elements enabling expression of a single gene in different organs, or allowing dynamic temporal changes corresponding to progressing phases of differentiation in a given cell type.
Collapse
Affiliation(s)
- Natalie Yivgi-Ohana
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Martin LJ, Boucher N, Brousseau C, Tremblay JJ. The orphan nuclear receptor NUR77 regulates hormone-induced StAR transcription in Leydig cells through cooperation with Ca2+/calmodulin-dependent protein kinase I. Mol Endocrinol 2008; 22:2021-37. [PMID: 18599618 DOI: 10.1210/me.2007-0370] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cholesterol transport in the mitochondrial membrane, an essential step of steroid biosynthesis, is mediated by a protein complex containing the steroidogenic acute regulatory (StAR) protein. The importance of this transporter is underscored by mutations in the human StAR gene that cause lipoid congenital adrenal hyperplasia, male pseudohermaphroditism, and adrenal insufficiency. StAR transcription in steroidogenic cells is hormonally regulated and involves several transcription factors. The nuclear receptor NUR77 is present in steroidogenic cells, and its expression is induced by hormones known to activate StAR expression. We have now established that StAR transcription in cAMP-stimulated Leydig cells requires de novo protein synthesis and involves NUR77. We found that cAMP-induced NUR77 expression precedes that of StAR both at the mRNA and protein levels in Leydig cells. In these cells, small interfering RNA-mediated NUR77 knockdown reduces cAMP-induced StAR expression. Chromatin immunoprecipitation assays revealed a cAMP-dependent increase in NUR77 recruitment to the proximal StAR promoter, whereas transient transfections in MA-10 Leydig cells confirmed that NUR77 can activate the StAR promoter and that this requires an element located at -95 bp. cAMP-induced StAR and NUR77 expression in Leydig cells was found to require a Ca2+/calmodulin-dependent protein kinase (CaMK)-dependent signaling pathway. Consistent with this, we show that within the testis, CaMKI is specifically expressed in Leydig cells. Finally, we report that CaMKI transcriptionally cooperates with NUR77, but not steroidogenic factor 1, to further enhance StAR promoter activity in Leydig cells. All together, our results implicate NUR77 as a mediator of cAMP action on StAR transcription in steroidogenic Leydig cells and identify a role for CaMKI in this process.
Collapse
Affiliation(s)
- Luc J Martin
- Reproduction, Perinatal and Child Health, Centre Hospitalier Universitaire of Quebec Research Centre, CHUL Room T1-49, 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2
| | | | | | | |
Collapse
|
23
|
Abstract
Steroid hormones are essential regulators of a vast number of physiological processes. The biosynthesis of these chemical messengers occurs in specialized steroidogenic tissues via a multi-step process that is catalyzed by members of the cytochrome P450 superfamily of monooxygenases and hydroxysteroid dehydrogenases. Though numerous signaling mediators, including cytokines and growth factors control steroidogenesis, trophic peptide hormones are the primary regulators of steroid hormone production. These peptide hormones activate a cAMP/cAMP-dependent kinase (PKA) signaling pathway, however, studies have shown that crosstalk between multiple signal transduction pathways and signaling molecules modulates optimal steroidogenic capacity. Sphingolipids such as ceramide, sphingosine, sphingosine-1-phosphate, sphingomyelin, and gangliosides have been shown to control the steroid hormone biosynthetic pathway at multiple levels, including regulating steroidogenic gene expression and activity as well as acting as second messengers in signaling cascades. In this review, we provide an overview of recent studies that have investigated the role of sphingolipids in adrenal, gonadal, and neural steroidogenesis.
Collapse
Affiliation(s)
- Natasha C Lucki
- School of Biology and Parker H, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332-0230, USA
| | | |
Collapse
|
24
|
Sugawara T, Fujimoto S. The potential function of steroid sulphatase activity in steroid production and steroidogenic acute regulatory protein expression. Biochem J 2004; 380:153-60. [PMID: 14969586 PMCID: PMC1224158 DOI: 10.1042/bj20031379] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Revised: 01/27/2004] [Accepted: 02/18/2004] [Indexed: 11/17/2022]
Abstract
The first step in the biosynthesis of steroid hormones is conversion of cholesterol into pregnenolone. StAR (steroidogenic acute regulatory) protein plays a crucial role in the intra-mitochondrial movement of cholesterol. STS (steroid sulphatase), which is present ubiquitously in mammalian tissues, including the placenta, adrenal gland, testis and ovary, desulphates a number of 3beta-hydroxysteroid sulphates, including cholesterol sulphate. The present study was designed to examine the effect of STS on StAR protein synthesis and steroidogenesis in cells. Steroidogenic activities of COS-1 cells that had been co-transfected with a vector for the cholesterol P450scc (cytochrome P450 side-chain-cleavage enzyme) system, named F2, a StAR expression vector (pStAR), and an STS expression vector (pSTS) were assayed. Whole-cell extracts were subjected to SDS/PAGE and then to Western blot analysis. pSTS co-expressed in COS-1 cells with F2 and pStAR increased pregnenolone synthesis 2-fold compared with that of co-expression with F2 and pStAR. Western blot analysis using COS-1 cells that had been co-transfected with pSTS, F2 and pStAR revealed that StAR protein levels increased, whereas STS and P450scc protein levels did not change. The amount of StAR protein translation products increased when pSTS was added to an in vitro transcription-translation reaction mixture. Pulse-chase experiments demonstrated that the 37 kDa StAR pre-protein disappeared significantly ( P <0.01) more slowly in COS-1 cells that had been transfected with pSTS than in COS-1 cells that had not been transfected with pSTS. The increase in StAR protein level is not a result of an increase in StAR gene expression, but is a result of both an increase in translation and a longer half-life of the 37 kDa pre-StAR protein. In conclusion, STS increases StAR protein expression level and stimulates steroid production.
Collapse
Affiliation(s)
- Teruo Sugawara
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, Kita-ku, Kita 15, Nishi 7, Sapporo 060-8638, Japan.
| | | |
Collapse
|
25
|
Val P, Lefrançois-Martinez AM, Veyssière G, Martinez A. SF-1 a key player in the development and differentiation of steroidogenic tissues. NUCLEAR RECEPTOR 2003; 1:8. [PMID: 14594453 PMCID: PMC240021 DOI: 10.1186/1478-1336-1-8] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Accepted: 09/18/2003] [Indexed: 12/16/2022]
Abstract
Since its discovery in the early 1990s, the orphan nuclear receptor SF-1 has been attributed a central role in the development and differentiation of steroidogenic tissues. SF-1 controls the expression of all the steroidogenic enzymes and cholesterol transporters required for steroidogenesis as well as the expression of steroidogenesis-stimulating hormones and their cognate receptors. SF-1 is also an essential regulator of genes involved in the sex determination cascade. The study of SF-1 null mice and of human mutants has been of great value to demonstrate the essential role of this factor in vivo, although the complete adrenal and gonadal agenesis in knock-out animals has impeded studies of its function as a transcriptional regulator. In particular, the role of SF-1 in the hormonal responsiveness of steroidogenic genes promoters is still a subject of debate. This extensive review takes into account recent data obtained from SF-1 haploinsufficient mice, pituitary-specific knock-outs and from transgenic mice experiments carried out with SF-1 target gene promoters. It also summarizes the pros and cons regarding the presumed role of SF-1 in cAMP signalling.
Collapse
Affiliation(s)
- Pierre Val
- UMR CNRS 6547, Physiologie Comparée et Endocrinologie Moléculaire, Université Blaise Pascal, Clermont II, Complexe Universitaire des Cézeaux, 24 avenue des Landais, 63177 Aubiere Cedex, France
| | - Anne-Marie Lefrançois-Martinez
- UMR CNRS 6547, Physiologie Comparée et Endocrinologie Moléculaire, Université Blaise Pascal, Clermont II, Complexe Universitaire des Cézeaux, 24 avenue des Landais, 63177 Aubiere Cedex, France
| | - Georges Veyssière
- UMR CNRS 6547, Physiologie Comparée et Endocrinologie Moléculaire, Université Blaise Pascal, Clermont II, Complexe Universitaire des Cézeaux, 24 avenue des Landais, 63177 Aubiere Cedex, France
| | - Antoine Martinez
- UMR CNRS 6547, Physiologie Comparée et Endocrinologie Moléculaire, Université Blaise Pascal, Clermont II, Complexe Universitaire des Cézeaux, 24 avenue des Landais, 63177 Aubiere Cedex, France
| |
Collapse
|
26
|
Jordan BK, Shen JHC, Olaso R, Ingraham HA, Vilain E. Wnt4 overexpression disrupts normal testicular vasculature and inhibits testosterone synthesis by repressing steroidogenic factor 1/beta-catenin synergy. Proc Natl Acad Sci U S A 2003; 100:10866-71. [PMID: 12949260 PMCID: PMC196894 DOI: 10.1073/pnas.1834480100] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genetic studies in mice suggest that Wnt4 signaling antagonizes expression of male hormones and effectively blocks male development in the female embryo. We recently identified an XY intersex patient carrying a chromosomal duplication of the WNT4 locus and proposed that this patient's feminization arises from an increased dosage of WNT4. To test this hypothesis, a transgenic mouse was generated with a large genomic P1 containing the human WNT4. Although a complete male to female intersex phenotype was not observed in WNT4 transgenic male mice, a dramatic reduction in steroidogenic acute regulatory protein was detected consistent with the marked reduction in serum and testicular androgen levels. Furthermore, a mild reduction of germ cells and a disorganized vascular system were observed in testes of WNT4 transgenic males. Consistent with these in vivo data, Wnt4 repressed steroidogenesis in adrenocortical and Leydig cell lines, as evidenced by reduced progesterone secretion and 3beta-hydroxysteroid dehydrogenase activity. In vitro studies showed that Wnt4 antagonizes the functional synergy observed between the major effector of the Wnt signaling pathway, beta-catenin and steroidogenic factor 1, and chromatin immunoprecipitation showed that Wnt4 attenuates recruitment of beta-catenin to the steroidogenic acute regulatory protein promoter. Our findings suggest a model in which Wnt4 acts as an anti-male factor by disrupting recruitment of beta-catenin at or near steroidogenic factor 1 binding sites present in multiple steroidogenic genes.
Collapse
Affiliation(s)
- Brian K Jordan
- Department of Human Genetics, University of California School of Medicine, Los Angeles, CA 90095-7088, USA
| | | | | | | | | |
Collapse
|
27
|
Wang Y, Newton DC, Miller TL, Teichert AM, Phillips MJ, Davidoff MS, Marsden PA. An alternative promoter of the human neuronal nitric oxide synthase gene is expressed specifically in Leydig cells. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:369-80. [PMID: 11786430 PMCID: PMC1867129 DOI: 10.1016/s0002-9440(10)64380-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neuronal nitric oxide synthase (nNOS) plays a modulatory role in the biology of a variety of neuroendocrine tissues and is especially relevant to gonadal function. We have previously reported the cloning and characterization of a variant of the nNOS protein, termed testis nNOS (TnNOS), the mRNA for which was restricted in expression to male gonadal tissues. To examine the cell-specificity of the testis-specific NOS regulatory regions we defined patterns of beta-galactosidase expression of an insertional transgene in which the reporter gene lacZ was under the transcriptional control of the human TnNOS promoter. beta-galactosidase activity was detected exclusively in the interstitial cells of the testis in transgenic mice. These cells also evidenced positive staining for nNOS protein and were identified as androgen-producing Leydig cells by staining with the Leydig cell marker, P(450)scc. Expression of the promoter was absent in cells of the seminiferous tubules, specifically germline cells of different stages and Sertoli cells. In contrast to the male gonad, beta-galactosidase activity was not detected in ovaries of adult female mice. Activity was also not evident in organs known to express full-length nNOS, such as skeletal muscle, kidney, or cerebellum. The same pattern of beta-galactosidase staining was observed in independent transgenic founders and was distinct from that observed for an endothelial NOS promoter/reporter transgene. In the testis of male adult eNOS promoter-reporter transgenic mice, beta-galactosidase activity was expressed only in endothelial cells of large- and medium-sized arterial blood vessels. Transcriptional activity of the human TnNOS promoter could not be detected in a variety of cell types, including Leydig cells, using episomal promoter-reporter constructs suggesting that a nuclear environment and higher order genomic complexity are required for appropriate promoter function. The restricted expression pattern of an nNOS variant in Leydig cells of the male gonad suggests an important role in the regulation of testosterone release and represents an intriguing model with which to dissect the molecular basis of Leydig cell-specific gene expression.
Collapse
Affiliation(s)
- Yang Wang
- Renal Division and Department of Medicine, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
28
|
Sewer MB, Waterman MR. Insights into the transcriptional regulation of steroidogenic enzymes and StAR. Rev Endocr Metab Disord 2001; 2:269-74. [PMID: 11705132 DOI: 10.1023/a:1011516532335] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- M B Sewer
- Vanderbilt University School of Medicine, Department of Biochemistry, 607 Light Hall, Nashville, Tennessee 37232-0146, USA
| | | |
Collapse
|
29
|
Lopez D, Nackley AC, Shea-Eaton W, Xue J, Schimmer BP, McLean MP. Effects of mutating different steroidogenic factor-1 protein regions on gene regulation. Endocrine 2001; 14:353-62. [PMID: 11444433 DOI: 10.1385/endo:14:3:353] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2000] [Revised: 01/17/2001] [Accepted: 01/17/2001] [Indexed: 11/11/2022]
Abstract
The involvement of cyclic adenosine monophosphate cAMP-dependent protein kinase A (PKA) in the regulation of the steroidogenic acute regulatory protein (StAR) and the high-density lipoprotein receptor (HDL-R) genes by steroidogenic factor-1 (SF-1) and cAMP were examined. Cotransfection studies carried out in Kin 8 cells, a Y1 cell line (mouse adrenal) with a mutation in the type I PKA regulatory subunit, demonstrated that an intact PKA is required for maximal activation and that SF-1 participates in cAMP regulation of these genes. Site-directed mutational analysis was performed to examine which SF-1 regions could be involved in SF-1 transcriptional activation of the StAR and HDL-R genes. SF-1 regions protein analyzed were amino acids Thr 60, Ser 203, Ser 431, Thr 462, and the activation function-2 domain (amino acids 449-462). Plasmids encoding each of the mutated SF-1 proteins were cotransfected with the StAR and HDL-R promoter constructs into human bladder carcinoma (HTB-9) cells in the presence or absence of dibutyryl cAMP. The results of these studies suggest that although SF-1 is required for optimal promoter response to cAMP, transcriptional activation of genes by SF-1 and cAMP are promoter dependent, perhaps resulting from gene-specific interactions of this transcription factor with other regulatory proteins.
Collapse
Affiliation(s)
- D Lopez
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, USA
| | | | | | | | | | | |
Collapse
|
30
|
Lopez D, Shea-Eaton W, McLean MP. Characterization of a steroidogenic factor-1-binding site found in promoter of sterol carrier protein-2 gene. Endocrine 2001; 14:253-61. [PMID: 11394644 DOI: 10.1385/endo:14:2:253] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Sterol carrier protein-2 (SCP2) is thought to mediate intracellular cholesterol transport in steroidogenic tissues. To elucidate the mechanism underlying the expression of this gene, a 300-bp fragment of the SCP2 promoter was cloned and analyzed for regulatory motifs. This promoter region contained a SF-1 binding motif, three activator protein-1 elements, an insulin response element, and a peroxisomal proliferator response element. The putative SF-1 binding region reacted with recombinant SF-1 DNA-binding domain in a mobility shift assay. The SCP2 promoter fragment was linked to a luciferase reporter gene and cotransfected in the presence or absence of SF-1 into HTB-9 cells. The results indicated that SF-1 was able to increase SCP2 promoter activity, an effect that was enhanced by cAMP. Similar results were obtained when the SCP2 promoter construct was cotransfected into Y1 cells. Cotransfection studies carried out in Kin 8 cells, a Y1 cell line with a mutation that prevents cAMP activation of PKA, revealed that a functional PKA is required for cAMP induction of SCP2 gene transcription. These results demonstrated that SF-1 confers cAMP responsiveness to the SCP2 promoter suggesting that SF-1 activation may be critical in regulation of this cholesterol transport protein.
Collapse
Affiliation(s)
- D Lopez
- Department of Obstetrics and Gynecology, University of South Florida, Tampa 33706, USA
| | | | | |
Collapse
|