1
|
Orta-Yilmaz B, Korkut A, Aydin Y. The impact of furan exposure on steroidogenesis in Leydig cells: cellular and molecular observations. Mol Biol Rep 2024; 51:1047. [PMID: 39388074 DOI: 10.1007/s11033-024-09954-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Furan is an organic compound that occurs as a result of heat treatment during the processing and cooking of many food products. Furthermore, the environment contains furan in tobacco smoke and vehicle exhaust gases, and it serves as an intermediate molecule in the synthesis of various pharmaceutical and chemical agents, pesticides, and stabilizers. Studies on the male reproductive system have not been able to elucidate the pathway through which furan exerts its negative effects. METHODS AND RESULTS In this study, the TM3 Leydig cell line was exposed to various furan concentrations (0.03, 0.3, and 3 mM) for 24 h. In order to assess the cytotoxic effects of furan on Leydig cells, we examined cell viability, cell proliferation, and lactate dehydrogenase enzyme levels. To investigate the detrimental effects of furan on testosterone biosynthesis, quantitative analyses were conducted on cAMP and testosterone levels, as well as the expression levels of key genes and transcription factors implicated in the steroidogenic pathway. The results indicate that furan inhibited the viability and proliferation of Leydig cells and enhanced the activity of lactate dehydrogenase. Leydig cells administered to furan exhibited notable reductions in cAMP and testosterone levels. Additionally, while the expression levels of steroidogenic genes were downregulated, significant changes were detected in the expression levels of the transcription factors responsible for the regulation of these genes. CONCLUSIONS Consequently, our findings suggest that furan exerts inhibitory effects on steroidogenesis in Leydig cells through multiple mechanisms, ultimately leading to infertility by inducing dysfunction in Leydig cells.
Collapse
Affiliation(s)
- Banu Orta-Yilmaz
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, Istanbul, 34134, Turkey.
| | - Ahu Korkut
- Department of Obstetrics and Gynecology, Perinatology Division, Antalya City Hospital, Antalya, Turkey
| | - Yasemin Aydin
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, Istanbul, 34134, Turkey
| |
Collapse
|
2
|
Sun W, Tian F, Pan H, Chang X, Xia M, Hu J, Wang Y, Li R, Li W, Yang M, Zhou Z. Flurochloridone induced abnormal spermatogenesis by damaging testicular Sertoli cells in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114163. [PMID: 36240522 DOI: 10.1016/j.ecoenv.2022.114163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/09/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Flurochloridone (FLC), a selective herbicide used on a global scale, has been reported to have male reproductive toxicity whose evidence is limited, but its mechanism remains unclear. The present study was conducted to systematically explore the male reproductive toxicity of FLC, including sperm quality, spermatogenesis, toxicity targets, and potential mechanisms. METHODS Male C57BL/6 mice aged 6-7 weeks received gavage administration of FLC (365/730 mg/kg/day) for 28 consecutive days. Then, the tissue and sperm of mice were collected for analysis. We measured the gonadosomatic index and analyzed sperm concentration, motility, malformation rate, and mitochondrial membrane potential (MMP). Spermatocyte immunofluorescence staining was performed to analyze meiosis. We also performed pathological staining on the testis and epididymis tissue and TUNEL staining, immunohistochemical analysis, and ultrastructural observation on the testicular tissue. RESULTS Results showed that FLC caused testicular weight reduction, dysfunction, and architectural damage in mice, but no significant adverse effect was found in the epididymis. The exposure interfered with spermatogonial proliferation and meiosis, affecting sperm concentration, motility, kinematic parameters, morphology, and MMP, decreasing sperm quality. Furthermore, mitochondrial damage and apoptosis of testicular Sertoli cells were observed in mice treated with FLC. CONCLUSION We found that FLC has significant adverse effects on spermatogonial proliferation and meiosis. Meanwhile, apoptosis and mitochondrial damage may be the potential mechanism of Sertoli cell damage. Our study demonstrated that FLC could induce testicular Sertoli cell damage, leading to abnormal spermatogenesis, which decreased sperm quality. The data provided references for the toxicity risk and research methods of FLC application in the environment.
Collapse
Affiliation(s)
- Weiqi Sun
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China; Shanghai Municipal Center for Disease Control and Prevention, Shanghai 200336, China
| | - Fang Tian
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Hongjie Pan
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Xiuli Chang
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China
| | - Minjie Xia
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Jingying Hu
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Yuzhu Wang
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Runsheng Li
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Weihua Li
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China
| | - Mingjun Yang
- Key Laboratory of Reproduction Regulation of National Health Commission (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai 200032, China.
| | - Zhijun Zhou
- School of Public Health/MOE Key Laboratory for Public Health Safety/ Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
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
|
4
|
Viger RS, de Mattos K, Tremblay JJ. Insights Into the Roles of GATA Factors in Mammalian Testis Development and the Control of Fetal Testis Gene Expression. Front Endocrinol (Lausanne) 2022; 13:902198. [PMID: 35692407 PMCID: PMC9178088 DOI: 10.3389/fendo.2022.902198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 12/28/2022] Open
Abstract
Defining how genes get turned on and off in a correct spatiotemporal manner is integral to our understanding of the development, differentiation, and function of different cell types in both health and disease. Testis development and subsequent male sex differentiation of the XY fetus are well-orchestrated processes that require an intricate network of cell-cell communication and hormonal signals that must be properly interpreted at the genomic level. Transcription factors are at the forefront for translating these signals into a coordinated genomic response. The GATA family of transcriptional regulators were first described as essential regulators of hematopoietic cell differentiation and heart morphogenesis but are now known to impact the development and function of a multitude of tissues and cell types. The mammalian testis is no exception where GATA factors play essential roles in directing the expression of genes crucial not only for testis differentiation but also testis function in the developing male fetus and later in adulthood. This minireview provides an overview of the current state of knowledge of GATA factors in the male gonad with a particular emphasis on their mechanisms of action in the control of testis development, gene expression in the fetal testis, testicular disease, and XY sex differentiation in humans.
Collapse
Affiliation(s)
- Robert S. Viger
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle and Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec—Université Laval, Quebec City, QC, Canada
| | - Karine de Mattos
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec—Université Laval, Quebec City, QC, Canada
| | - Jacques J. Tremblay
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle and Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec—Université Laval, Quebec City, QC, Canada
| |
Collapse
|
5
|
Liu H, Fan M, Fu X, Chen Y, Ye M, Guo H. Simultaneous Determination of Prostaglandin and Hormones in Excreta of Trogopterus xanthipes. J Chromatogr Sci 2021; 58:542-548. [PMID: 32405648 DOI: 10.1093/chromsci/bmaa017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/08/2020] [Accepted: 04/04/2020] [Indexed: 11/12/2022]
Abstract
The excreta of Trogopterus xanthipes (also called Wulingzhi in Chinese, WLZ) is a well-known traditional Chinese medicine used for the treatment of irregular menstruation in clinic. Few reports are available on the chemical profiling of WLZ. In this work, qualitative and quantitative analyses of endogenous prostaglandin and hormones in WLZ were performed using UHPLC-orbitrap-MSn. In total, 48 compounds were identified in urine of T. xanthipes. Furthermore, the contents of four target compounds were simultaneously quantitated in 20 batches of samples by UPLC-MS/MS. The quantitative method showed a good linear correlation (R > 0.995) in a wide range for each compound. The method had a high sensitivity with LOD (0.5-1.0 ng/mL) and LOQ (1.0-2.5 ng/mL). The intra- and inter-day precisions were < 9.17 (RSD %), and repeatability and stability were < 6.14 (RSD %). The recovery of the analytes varied between 85.8% and 97.3% at three different concentrations. The present integrated qualitative and quantitative assessment of WLZ provides an evaluation strategy to assess the constituent in traditional Chinese medicine.
Collapse
Affiliation(s)
- Haolong Liu
- School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, 25 Kexueyuan Road, Beijing 102206, China
| | - Miaoxuan Fan
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, 25 Kexueyuan Road, Beijing 102206, China
| | - Xintong Fu
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, 25 Kexueyuan Road, Beijing 102206, China
| | - Yougen Chen
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, 25 Kexueyuan Road, Beijing 102206, China
| | - Min Ye
- School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
| | - Hongzhu Guo
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, 25 Kexueyuan Road, Beijing 102206, China
| |
Collapse
|
6
|
DeWitt NA, Whirledge S, Kallen AN. Updates on molecular and environmental determinants of luteal progesterone production. Mol Cell Endocrinol 2020; 515:110930. [PMID: 32610113 PMCID: PMC7484338 DOI: 10.1016/j.mce.2020.110930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
Progesterone, a critical hormone in reproduction, is a key sex steroid in the establishment and maintenance of early pregnancy and serves as an intermediary for synthesis of other steroid hormones. Progesterone production from the corpus luteum is a tightly regulated process which is stimulated and maintained by multiple factors, both systemic and local. Multiple regulatory systems, including classic mediators of gonadotropin stimulation such as the cAMP/PKA pathway and TGFβ-mediated signaling pathways, as well as local production of hormonal factors, exist to promote granulosa cell function and physiological fine-tuning of progesterone levels. In this manuscript, we provide an updated narrative review of the known mediators of human luteal progesterone and highlight new observations regarding this important process, focusing on studies published within the last five years. We will also review recent evidence suggesting that this complex system of progesterone production is sensitive to disruption by exogenous environmental chemicals that can mimic or interfere with the activities of endogenous hormones.
Collapse
Affiliation(s)
- Natalie A DeWitt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Shannon Whirledge
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Amanda N Kallen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Bouchard MF, Bergeron F, Grenier Delaney J, Harvey LM, Viger RS. In Vivo Ablation of the Conserved GATA-Binding Motif in the Amh Promoter Impairs Amh Expression in the Male Mouse. Endocrinology 2019; 160:817-826. [PMID: 30759208 PMCID: PMC6426834 DOI: 10.1210/en.2019-00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 02/08/2019] [Indexed: 12/23/2022]
Abstract
GATA4 is an essential transcriptional regulator required for gonadal development, differentiation, and function. In the developing testis, proposed GATA4-regulated genes include steroidogenic factor 1 (Nr5a1), SRY-related HMG box 9 (Sox9), and anti-Müllerian hormone (Amh). Although some of these genes have been validated as genuine GATA4 targets, it remains unclear whether GATA4 is a direct regulator of endogenous Amh transcription. We used a CRISPR/Cas9-based approach to specifically inactivate or delete the sole GATA-binding motif of the proximal mouse Amh promoter. AMH mRNA and protein levels were assessed at developmental time points corresponding to elevated AMH levels: fetal and neonate testes in males and adult ovaries in females. In males, loss of GATA binding to the Amh promoter significantly reduced Amh expression. Although the loss of GATA binding did not block the initiation of Amh transcription, AMH mRNA and protein levels failed to upregulate in the developing fetal and neonate testis. Interestingly, adult male mice presented no anatomical anomalies and had no evidence of retained Müllerian duct structures, suggesting that AMH levels, although markedly reduced, were sufficient to masculinize the male embryo. In contrast to males, GATA binding to the Amh promoter was dispensable for Amh expression in the adult ovary. These results provide conclusive evidence that in males, GATA4 is a positive modulator of Amh expression that works in concert with other key transcription factors to ensure that the Amh gene is sufficiently expressed in a correct spatiotemporal manner during fetal and prepubertal testis development.
Collapse
Affiliation(s)
- Marie France Bouchard
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Francis Bergeron
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Jasmine Grenier Delaney
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Louis-Mathieu Harvey
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Robert S Viger
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
- Department of Obstetrics, Gynecology, and Reproduction, Université Laval, Quebec, Quebec, Canada
- Correspondence: Robert S. Viger, PhD, Reproduction, Mother and Child Health, Room T3-67, Centre de Recherche du CHU de Québec–Université Laval, 2705 Laurier Boulevard, Quebec, Quebec G1V 4G2, Canada. E-mail:
| |
Collapse
|
8
|
Bergeron F, Boulende Sab A, Bouchard MF, Taniguchi H, Souchkova O, Brousseau C, Tremblay JJ, Pilon N, Viger RS. Phosphorylation of GATA4 serine 105 but not serine 261 is required for testosterone production in the male mouse. Andrology 2019; 7:357-372. [PMID: 30793514 DOI: 10.1111/andr.12601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND GATA4 is a transcription factor essential for male sex determination, testicular differentiation during fetal development, and male fertility in the adult. GATA4 exerts part of its function by regulating multiple genes in the steroidogenic enzyme pathway. In spite of these crucial roles, how the activity of this factor is regulated remains unclear. OBJECTIVES Studies in gonadal cell lines have shown that GATA4 is phosphorylated on at least two serine residues-serine 105 (S105) and serine 261 (S261)-and that this phosphorylation is important for GATA4 activity. The objective of the present study is to characterize the endogenous role of GATA4 S105 and S261 phosphorylation in the mouse testis. MATERIALS AND METHODS We examined both previously described GATA4 S105A mice and a novel GATA4 S261A knock-in mouse that we generated by CRISPR/Cas9 gene editing. The male phenotype of the mutants was characterized by assessing androgen-dependent organ weights, hormonal profiles, and expression of multiple testicular target genes using standard biochemical and molecular biology techniques. RESULTS The fecundity of crosses between GATA4 S105A mice was reduced but without a change in sex ratio. The weight of androgen-dependent organs was smaller when compared to wild-type controls. Plasma testosterone levels showed a 70% decrease in adult GATA4 S105A males. This decrease was associated with a reduction in Cyp11a1, Cyp17a1, and Hsd17b3 expression. GATA4 S261A mice were viable and testis morphology appeared normal. Testosterone production and steroidogenic enzyme expression were not altered in GATA4 S261A males. DISCUSSION AND CONCLUSION Our analysis showed that blocking GATA4 S105 phosphorylation is associated with decreased androgen production in males. In contrast, S261 phosphorylation by itself is dispensable for GATA4 function. These results confirm that endogenous GATA4 action is essential for normal steroid production in males and that this activity requires phosphorylation on at least one serine residue.
Collapse
Affiliation(s)
- F Bergeron
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Quebec, QC, Canada
| | - A Boulende Sab
- Département des Sciences Biologiques and Centre d'excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - M F Bouchard
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Quebec, QC, Canada
| | - H Taniguchi
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - O Souchkova
- Département des Sciences Biologiques and Centre d'excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - C Brousseau
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Quebec, QC, Canada
| | - J J Tremblay
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Quebec, QC, Canada.,Department of Obstetrics, Gynecology, and Reproduction, Université Laval, Quebec, QC, Canada
| | - N Pilon
- Département des Sciences Biologiques and Centre d'excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montreal, QC, Canada
| | - R S Viger
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Quebec, QC, Canada.,Department of Obstetrics, Gynecology, and Reproduction, Université Laval, Quebec, QC, Canada
| |
Collapse
|
9
|
Bennett-Toomey J, Stocco C. GATA Regulation and Function During the Ovarian Life Cycle. VITAMINS AND HORMONES 2018; 107:193-225. [PMID: 29544631 DOI: 10.1016/bs.vh.2018.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
GATA4 and GATA6 are the sole GATA factors expressed in the ovary during embryonic development and adulthood. Up today, GATA4 and GATA6 are the only transcription factors that have been conditionally deleted during ovarian development and at each major stage of follicle maturation. The evidence from these transgenic mice revealed that GATA4 and GATA6 are crucial for follicles assembly, granulosa cell differentiation, postnatal follicle growth, and luteinization. Thus, conditional knockdown of both factors in the granulosa cells at any stage of development leads to female infertility. GATA targets impacting female reproduction include genes involved in steroidogenesis, hormone signaling, ovarian hormones, extracellular matrix organization, and apoptosis/cell division.
Collapse
Affiliation(s)
| | - Carlos Stocco
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
10
|
Laissue P. The molecular complexity of primary ovarian insufficiency aetiology and the use of massively parallel sequencing. Mol Cell Endocrinol 2018; 460:170-180. [PMID: 28743519 DOI: 10.1016/j.mce.2017.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 11/28/2022]
Abstract
Primary ovarian insufficiency (POI) is a frequently occurring pathology, leading to infertility. Genetic anomalies have been described in POI and mutations in numerous genes have been definitively related to the pathogenesis of the disease. Some studies based on next generation sequencing (NGS) have been successfully undertaken as they have led to identify new mutations associated with POI aetiology. The purpose of this review is to present the most relevant molecules involved in diverse complex pathways, which may contribute towards POI. The main genes participating in bipotential gonad formation, sex determination, meiosis, folliculogenesis and ovulation are described to enable understanding how they may be considered putative candidates involved in POI. Considerations regarding NGS technical aspects such as design and data interpretation are mentioned. Successful NGS initiatives used for POI studying and future challenges are also discussed.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
11
|
Penny GM, Cochran RB, Pihlajoki M, Kyrönlahti A, Schrade A, Häkkinen M, Toppari J, Heikinheimo M, Wilson DB. Probing GATA factor function in mouse Leydig cells via testicular injection of adenoviral vectors. Reproduction 2017; 154:455-467. [PMID: 28710293 PMCID: PMC5589507 DOI: 10.1530/rep-17-0311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/09/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022]
Abstract
Testicular Leydig cells produce androgens essential for proper male reproductive development and fertility. Here, we describe a new Leydig cell ablation model based on Cre/Lox recombination of mouse Gata4 and Gata6, two genes implicated in the transcriptional regulation of steroidogenesis. The testicular interstitium of adult Gata4flox/flox ; Gata6flox/flox mice was injected with adenoviral vectors encoding Cre + GFP (Ad-Cre-IRES-GFP) or GFP alone (Ad-GFP). The vectors efficiently and selectively transduced Leydig cells, as evidenced by GFP reporter expression. Three days after Ad-Cre-IRES-GFP injection, expression of androgen biosynthetic genes (Hsd3b1, Cyp17a1 and Hsd17b3) was reduced, whereas expression of another Leydig cell marker, Insl3, was unchanged. Six days after Ad-Cre-IRES-GFP treatment, the testicular interstitium was devoid of Leydig cells, and there was a concomitant loss of all Leydig cell markers. Chromatin condensation, nuclear fragmentation, mitochondrial swelling, and other ultrastructural changes were evident in the degenerating Leydig cells. Liquid chromatography-tandem mass spectrometry demonstrated reduced levels of androstenedione and testosterone in testes from mice injected with Ad-Cre-IRES-GFP. Late effects of treatment included testicular atrophy, infertility and the accumulation of lymphoid cells in the testicular interstitium. We conclude that adenoviral-mediated gene delivery is an expeditious way to probe Leydig cell function in vivo Our findings reinforce the notion that GATA factors are key regulators of steroidogenesis and testicular somatic cell survival.Free Finnish abstract: A Finnish translation of this abstract is freely available at http://www.reproduction-online.org/content/154/4/455/suppl/DC2.
Collapse
Affiliation(s)
- Gervette M Penny
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Rebecca B Cochran
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Marjut Pihlajoki
- Children's HospitalUniversity of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Kyrönlahti
- Children's HospitalUniversity of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anja Schrade
- Children's HospitalUniversity of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Merja Häkkinen
- University of Eastern FinlandSchool of Pharmacy, Kuopio, Finland
| | - Jorma Toppari
- Department of PhysiologyInstitute of Biomedicine, University of Turku and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Markku Heikinheimo
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
- Children's HospitalUniversity of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David B Wilson
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
- Department of Developmental BiologyWashington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Ye L, Li X, Li L, Chen H, Ge RS. Insights into the Development of the Adult Leydig Cell Lineage from Stem Leydig Cells. Front Physiol 2017; 8:430. [PMID: 28701961 PMCID: PMC5487449 DOI: 10.3389/fphys.2017.00430] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/06/2017] [Indexed: 02/06/2023] Open
Abstract
Adult Leydig cells (ALCs) are the steroidogenic cells in the testes that produce testosterone. ALCs develop postnatally from a pool of stem cells, referred to as stem Leydig cells (SLCs). SLCs are spindle-shaped cells that lack steroidogenic cell markers, including luteinizing hormone (LH) receptor and 3β-hydroxysteroid dehydrogenase. The commitment of SLCs into the progenitor Leydig cells (PLCs), the first stage in the lineage, requires growth factors, including Dessert Hedgehog (DHH) and platelet-derived growth factor-AA. PLCs are still spindle-shaped, but become steroidogenic and produce mainly androsterone. The next transition in the lineage is from PLC to the immature Leydig cell (ILC). This transition requires LH, DHH, and androgen. ILCs are ovoid cells that are competent for producing a different form of androgen, androstanediol. The final stage in the developmental lineage is ALC. The transition to ALC involves the reduced expression of 5α-reductase 1, a step that is necessary to make the cells to produce testosterone as the final product. The transitions along the Leydig cell lineage are associated with the progressive down-regulation of the proliferative activity, and the up-regulation of steroidogenic capacity, with each step requiring unique regulatory signaling.
Collapse
Affiliation(s)
- Leping Ye
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Xiaoheng Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Haolin Chen
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| | - Ren-Shan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou, China
| |
Collapse
|
13
|
Guo Y, Yu P, Liu Z, Maimaiti Y, Chen C, Zhang Y, Yin X, Wang S, Liu C, Huang T. Prognostic and clinicopathological value of GATA binding protein 3 in breast cancer: A systematic review and meta-analysis. PLoS One 2017; 12:e0174843. [PMID: 28394898 PMCID: PMC5386271 DOI: 10.1371/journal.pone.0174843] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
The potential prognostic value of GATA binding protein 3 (GATA3) in breast cancer has recently increased, although the evidence is inconclusive. This meta-analysis of 10 articles involving 5,080 breast cancer patients explored the prognostic and clinicopathological value of GATA3 in breast cancer. Time to tumor progression (TTP) and overall survival (OS) were primary endpoints. Pooled hazard ratio (HR), pooled risk ratio (RR), and 95% confidence interval (CI) were calculated to evaluate the association between GATA3, prognosis, and clinicopathological parameters. High GATA3 expression predicts breast cancer, with a HR (HR = 0.671; 95% CI = 0.475–0.947; P = 0.023) of TTP, but is not associated with OS (HR = 0.889; 95% CI = 0.789–1.001; P = 0.052). GATA3 overexpression is associated with positive ER (RR = 3.155; 95% CI = 1.680–5.923; P = 0.000), positive PR (RR = 3.949; 95% CI = 1.567–9.954, P = 0.004), lower nuclear grade (RR = 0.435; 95% CI = 0.369–0.514; P = 0.000), and smaller tumor size (RR = 0.816; 95% CI = 0.709–0.940; P = 0.005). High GATA3 expression may predict TTP in breast cancer, and such patients may show better clinicopathological features.
Collapse
Affiliation(s)
- Yawen Guo
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Pan Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Zeming Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Yusufu Maimaiti
- Department of General Surgery, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chen Chen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Yunke Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Xingjie Yin
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Shan Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
- * E-mail: (TH); (CL)
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science And Technology, Wuhan, China
- * E-mail: (TH); (CL)
| |
Collapse
|
14
|
Gustin SE, Stringer JM, Hogg K, Sinclair AH, Western PS. FGF9, activin and TGFβ promote testicular characteristics in an XX gonad organ culture model. Reproduction 2016; 152:529-43. [DOI: 10.1530/rep-16-0293] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/05/2016] [Indexed: 12/29/2022]
Abstract
Testis development is dependent on the key sex-determining factors SRY and SOX9, which activate the essential ligand FGF9. Although FGF9 plays a central role in testis development, it is unable to induce testis formation on its own. However, other growth factors, including activins and TGFβs, also present testis during testis formation. In this study, we investigated the potential of FGF9 combined with activin and TGFβ to induce testis development in cultured XX gonads. Our data demonstrated differing individual and combined abilities of FGF9, activin and TGFβ to promote supporting cell proliferation, Sertoli cell development and male germ line differentiation in cultured XX gonads. FGF9 promoted proliferation of supporting cells in XX foetal gonads at rates similar to those observed in vivo during testis cord formation in XY gonads but was insufficient to initiate testis development. However, when FGF9, activin and TGFβ were combined, aspects of testicular development were induced, including the expression of Sox9, morphological reorganisation of the gonad and deposition of laminin around germ cells. Enhancing β-catenin activity diminished the testis-promoting activities of the combined growth factors. The male promoting activity of FGF9 and the combined growth factors directly or indirectly extended to the germ line, in which a mixed phenotype was observed. FGF9 and the combined growth factors promoted male germ line development, including mitotic arrest, but expression of pluripotency genes was maintained, rather than being repressed. Together, our data provide evidence that combined signalling by FGF9, activin and TGFβ can induce testicular characteristics in XX gonads.
Collapse
|
15
|
Figueiredo AFA, França LR, Hess RA, Costa GMJ. Sertoli cells are capable of proliferation into adulthood in the transition region between the seminiferous tubules and the rete testis in Wistar rats. Cell Cycle 2016; 15:2486-96. [PMID: 27420022 DOI: 10.1080/15384101.2016.1207835] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Sertoli cells (SCs) play a crucial role in testis differentiation, development and function, determining the magnitude of sperm production in sexually mature animals. For over 40 years, it has been considered that these key testis somatic cells stop dividing during early pre-pubertal phase, between around 10 to 20 days after birth respectively in mice and rats, being after that under physiological conditions a stable and terminally differentiated population. However, evidences from the literature are challenging this dogma. In the present study, using several important functional markers (Ki-67, BrdU, p27, GATA-4, Androgen Receptor), we investigated the SC differentiation status in 36 days old and adult Wistar rats, focusing mainly in the transition region (TR) between the seminiferous tubules (ST) and the rete testis. Our results showed that SCs in TR remain undifferentiated for a longer period and, although at a lesser degree, even in adult rats proliferating SCs were observed in this region. Therefore, these findings suggest that, different from the other ST regions investigated, SCs residing in the TR exhibit a distinct functional phenotype. These undifferentiated SCs may compose a subpopulation of SC progenitors that reside in a specific microenvironment capable of growing the ST length if needed from this particular testis region. Moreover, our findings demonstrate an important aspect of testis function in mammals and opens new venues for other experimental approaches to the investigation of SC physiology, spermatogenesis progression and testis growth. Besides that, the TR may represent an important site for pathophysiological investigations and cellular interactions in the testis.
Collapse
Affiliation(s)
- A F A Figueiredo
- a Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - L R França
- a Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , MG , Brazil.,b National Institute of Amazonian Research (INPA/Manaus), National Institute of Amazonian Research (INPA) , Manaus , AM , Brazil
| | - R A Hess
- c Department of Comparative Biosciences , University of Illinois , Urbana Champaign , IL , USA
| | - G M J Costa
- a Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| |
Collapse
|
16
|
Schrade A, Kyrönlahti A, Akinrinade O, Pihlajoki M, Fischer S, Rodriguez VM, Otte K, Velagapudi V, Toppari J, Wilson DB, Heikinheimo M. GATA4 Regulates Blood-Testis Barrier Function and Lactate Metabolism in Mouse Sertoli Cells. Endocrinology 2016; 157:2416-31. [PMID: 26974005 PMCID: PMC4891789 DOI: 10.1210/en.2015-1927] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Conditional deletion of Gata4 in Sertoli cells (SCs) of adult mice has been shown to increase permeability of the blood-testis barrier (BTB) and disrupt spermatogenesis. To gain insight into the molecular underpinnings of these phenotypic abnormalities, we assessed the impact of Gata4 gene silencing in cell culture models. Microarray hybridization identified genes dysregulated by siRNA-mediated inhibition of Gata4 in TM4 cells, an immortalized mouse SC line. Differentially expressed genes were validated by quantitative RT-PCR analysis of primary cultures of Gata4(flox/flox) mouse SCs that had been subjected to cre-mediated recombination in vitro. Depletion of GATA4 in TM4 cells and primary SCs was associated with altered expression of genes involved in key facets of BTB maintenance, including tight/adherens junction formation (Tjp1, Cldn12, Vcl, Tnc, Csk) and extracellular matrix reorganization (Lamc1, Col4a1, Col4a5, Mmp10, Mmp23, Timp2). Western blotting and immunocytochemistry demonstrated reduced levels of tight junction protein-1, a prototypical tight junction protein, in GATA4-depleted cells. These changes were accompanied by a loss of morphologically recognizable junctional complexes and a decline in epithelial membrane resistance. Furthermore, Gata4 gene silencing was associated with altered expression of Hk1, Gpi1, Pfkp, Pgam1, Gls2, Pdk3, Pkd4, and Ldhb, genes regulating the production of lactate, a key nutrient that SCs provide to developing germ cells. Comprehensive metabolomic profiling demonstrated impaired lactate production in GATA4-deficient SCs. We conclude that GATA4 plays a pivotal role in the regulation of BTB function and lactate metabolism in mouse SCs.
Collapse
Affiliation(s)
- Anja Schrade
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Antti Kyrönlahti
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Oyediran Akinrinade
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Marjut Pihlajoki
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Simon Fischer
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Verena Martinez Rodriguez
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Kerstin Otte
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Vidya Velagapudi
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Jorma Toppari
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - David B Wilson
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| | - Markku Heikinheimo
- Children's Hospital (A.S., A.K., O.A., M.P., M.H.), University of Helsinki and Helsinki University Central Hospital, Helsinki 00014, Finland; Institute of Applied Biotechnology (S.F., K.O.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, Turku 20520, Finland; and Departments of Pediatrics (A.S., V.M.R., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University, St Louis, Missouri 63110
| |
Collapse
|
17
|
Pihlajoki M, Färkkilä A, Soini T, Heikinheimo M, Wilson DB. GATA factors in endocrine neoplasia. Mol Cell Endocrinol 2016; 421:2-17. [PMID: 26027919 PMCID: PMC4662929 DOI: 10.1016/j.mce.2015.05.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 04/26/2015] [Accepted: 05/09/2015] [Indexed: 02/07/2023]
Abstract
GATA transcription factors are structurally-related zinc finger proteins that recognize the consensus DNA sequence WGATAA (the GATA motif), an essential cis-acting element in the promoters and enhancers of many genes. These transcription factors regulate cell fate specification and differentiation in a wide array of tissues. As demonstrated by genetic analyses of mice and humans, GATA factors play pivotal roles in the development, homeostasis, and function of several endocrine organs including the adrenal cortex, ovary, pancreas, parathyroid, pituitary, and testis. Additionally, GATA factors have been shown to be mutated, overexpressed, or underexpressed in a variety of endocrine tumors (e.g., adrenocortical neoplasms, parathyroid tumors, pituitary adenomas, and sex cord stromal tumors). Emerging evidence suggests that GATA factors play a direct role in the initiation, proliferation, or propagation of certain endocrine tumors via modulation of key developmental signaling pathways implicated in oncogenesis, such as the WNT/β-catenin and TGFβ pathways. Altered expression or function of GATA factors can also affect the metabolism, ploidy, and invasiveness of tumor cells. This article provides an overview of the role of GATA factors in endocrine neoplasms. Relevant animal models are highlighted.
Collapse
Affiliation(s)
- Marjut Pihlajoki
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290 Helsinki, Finland
| | - Anniina Färkkilä
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290 Helsinki, Finland; Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, 00290 Helsinki, Finland
| | - Tea Soini
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290 Helsinki, Finland
| | - Markku Heikinheimo
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290 Helsinki, Finland; Department of Pediatrics, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David B Wilson
- Department of Pediatrics, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Suzuki H, Kanai-Azuma M, Kanai Y. From Sex Determination to Initial Folliculogenesis in Mammalian Ovaries: Morphogenetic Waves along the Anteroposterior and Dorsoventral Axes. Sex Dev 2015; 9:190-204. [DOI: 10.1159/000440689] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2015] [Indexed: 11/19/2022] Open
|
19
|
George RM, Hahn KL, Rawls A, Viger RS, Wilson-Rawls J. Notch signaling represses GATA4-induced expression of genes involved in steroid biosynthesis. Reproduction 2015; 150:383-94. [PMID: 26183893 DOI: 10.1530/rep-15-0226] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022]
Abstract
Notch2 and Notch3 and genes of the Notch signaling network are dynamically expressed in developing follicles, where they are essential for granulosa cell proliferation and meiotic maturation. Notch receptors, ligands, and downstream effector genes are also expressed in testicular Leydig cells, predicting a potential role in regulating steroidogenesis. In this study, we sought to determine if Notch signaling in small follicles regulates the proliferation response of granulosa cells to FSH and represses the up-regulation steroidogenic gene expression that occurs in response to FSH as the follicle grows. Inhibition of Notch signaling in small preantral follicles led to the up-regulation of the expression of genes in the steroid biosynthetic pathway. Similarly, progesterone secretion by MA-10 Leydig cells was significantly inhibited by constitutively active Notch. Together, these data indicated that Notch signaling inhibits steroidogenesis. GATA4 has been shown to be a positive regulator of steroidogenic genes, including STAR protein, P450 aromatase, and 3B-hydroxysteroid dehydrogenase. We observed that Notch downstream effectors HEY1, HEY2, and HEYL are able to differentially regulate these GATA4-dependent promoters. These data are supported by the presence of HEY/HES binding sites in these promoters. These studies indicate that Notch signaling has a role in the complex regulation of the steroidogenic pathway.
Collapse
Affiliation(s)
- Rajani M George
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Katherine L Hahn
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Alan Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Robert S Viger
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4 School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Jeanne Wilson-Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| |
Collapse
|
20
|
Röhrig T, Pihlajoki M, Ziegler R, Cochran RS, Schrade A, Schillebeeckx M, Mitra RD, Heikinheimo M, Wilson DB. Toying with fate: Redirecting the differentiation of adrenocortical progenitor cells into gonadal-like tissue. Mol Cell Endocrinol 2015; 408:165-77. [PMID: 25498963 PMCID: PMC4417465 DOI: 10.1016/j.mce.2014.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/30/2014] [Accepted: 12/01/2014] [Indexed: 01/07/2023]
Abstract
Cell fate decisions are integral to zonation and remodeling of the adrenal cortex. Animal models exhibiting ectopic differentiation of gonadal-like cells in the adrenal cortex can shed light on the molecular mechanisms regulating steroidogenic cell fate. In one such model, prepubertal gonadectomy (GDX) of mice triggers the formation of adrenocortical neoplasms that resemble luteinized ovarian stroma. Transcriptomic analysis and genome-wide DNA methylation mapping have identified genetic and epigenetic markers of GDX-induced adrenocortical neoplasia. Members of the GATA transcription factor family have emerged as key regulators of cell fate in this model. Expression of Gata4 is pivotal for the accumulation of gonadal-like cells in the adrenal glands of gonadectomized mice, whereas expression of Gata6 limits the spontaneous and GDX-induced differentiation of gonadal-like cells in the adrenal cortex. Additionally, Gata6 is essential for proper development of the adrenal X-zone, a layer analogous to the fetal zone of the human adrenal cortex. The relevance of these observations to developmental signaling pathways in the adrenal cortex, to other animal models of altered adrenocortical cell fate, and to human diseases is discussed.
Collapse
Affiliation(s)
- Theresa Röhrig
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim - University of Applied Sciences, Mannheim 68163, Germany
| | - Marjut Pihlajoki
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Ricarda Ziegler
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim - University of Applied Sciences, Mannheim 68163, Germany
| | - Rebecca S Cochran
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Anja Schrade
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Maximiliaan Schillebeeckx
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Markku Heikinheimo
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - David B Wilson
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA.
| |
Collapse
|
21
|
Heikinheimo M, Pihlajoki M, Schrade A, Kyrönlahti A, Wilson DB. Testicular steroidogenic cells to the rescue. Endocrinology 2015; 156:1616-9. [PMID: 25886071 DOI: 10.1210/en.2015-1222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Markku Heikinheimo
- Departments of Pediatrics and Developmental Biology (M.H., D.B.W.), Washington University School of Medicine and St Louis Children's Hospital, St Louis, Missouri 63110; and Children's Hospital (M.H., M.P., A.S., A.K.), University of Helsinki and Helsinki Central Hospital, 00290 Helsinki, Finland
| | | | | | | | | |
Collapse
|
22
|
Padua MB, Jiang T, Morse DA, Fox SC, Hatch HM, Tevosian SG. Combined loss of the GATA4 and GATA6 transcription factors in male mice disrupts testicular development and confers adrenal-like function in the testes. Endocrinology 2015; 156:1873-86. [PMID: 25668066 PMCID: PMC4398756 DOI: 10.1210/en.2014-1907] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The roles of the GATA4 and GATA6 transcription factors in testis development were examined by simultaneously ablating Gata4 and Gata6 with Sf1Cre (Nr5a1Cre). The deletion of both genes resulted in a striking testicular phenotype. Embryonic Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) (conditional double mutant) testes were smaller than control organs and contained irregular testis cords and fewer gonocytes. Gene expression analysis revealed significant down-regulation of Dmrt1 and Mvh. Surprisingly, Amh expression was strongly up-regulated and remained high beyond postnatal day 7, when it is normally extinguished. Neither DMRT1 nor GATA1 was detected in the Sertoli cells of the mutant postnatal testes. Furthermore, the expression of the steroidogenic genes Star, Cyp11a1, Hsd3b1, and Hsd17b3 was low throughout embryogenesis. Immunohistochemical analysis revealed a prominent reduction in cytochrome P450 side-chain cleavage enzyme (CYP11A1)- and 3β-hydroxysteroid dehydrogenase-positive (3βHSD) cells, with few 17α-hydroxylase/17,20 lyase-positive (CYP17A1) cells present. In contrast, in postnatal Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) testes, the expression of the steroidogenic markers Star, Cyp11a1, and Hsd3b6 was increased, but a dramatic down-regulation of Hsd17b3, which is required for testosterone synthesis, was observed. The genes encoding adrenal enzymes Cyp21a1, Cyp11b1, Cyp11b2, and Mcr2 were strongly up-regulated, and clusters containing numerous CYP21A2-positive cells were localized in the interstitium. These data suggest a lack of testis functionality, with a loss of normal steroidogenic testis function, concomitant with an expansion of the adrenal-like cell population in postnatal conditional double mutant testes. Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) animals of both sexes lack adrenal glands; however, despite this deficiency, males are viable in contrast to the females of the same genotype, which die shortly after birth.
Collapse
Affiliation(s)
- Maria B Padua
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610
| | | | | | | | | | | |
Collapse
|
23
|
Schrade A, Kyrönlahti A, Akinrinade O, Pihlajoki M, Häkkinen M, Fischer S, Alastalo TP, Velagapudi V, Toppari J, Wilson DB, Heikinheimo M. GATA4 is a key regulator of steroidogenesis and glycolysis in mouse Leydig cells. Endocrinology 2015; 156:1860-72. [PMID: 25668067 PMCID: PMC4398762 DOI: 10.1210/en.2014-1931] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transcription factor GATA4 is expressed in somatic cells of the mammalian testis. Gene targeting studies in mice have shown that GATA4 is essential for proper differentiation and function of Sertoli cells. The role of GATA4 in Leydig cell development, however, remains controversial, because targeted mutagenesis experiments in mice have not shown a consistent phenotype, possibly due to context-dependent effects or compensatory responses. We therefore undertook a reductionist approach to study the function of GATA4 in Leydig cells. Using microarray analysis and quantitative RT-PCR, we identified a set of genes that are down-regulated or up-regulated after small interfering RNA (siRNA)-mediated silencing of Gata4 in the murine Leydig tumor cell line mLTC-1. These same genes were dysregulated when primary cultures of Gata4(flox/flox) adult Leydig cells were subjected to adenovirus-mediated cre-lox recombination in vitro. Among the down-regulated genes were enzymes of the androgen biosynthetic pathway (Cyp11a1, Hsd3b1, Cyp17a1, and Srd5a). Silencing of Gata4 expression in mLTC-1 cells was accompanied by reduced production of sex steroid precursors, as documented by mass spectrometric analysis. Comprehensive metabolomic analysis of GATA4-deficient mLTC-1 cells showed alteration of other metabolic pathways, notably glycolysis. GATA4-depleted mLTC-1 cells had reduced expression of glycolytic genes (Hk1, Gpi1, Pfkp, and Pgam1), lower intracellular levels of ATP, and increased extracellular levels of glucose. Our findings suggest that GATA4 plays a pivotal role in Leydig cell function and provide novel insights into metabolic regulation in this cell type.
Collapse
Affiliation(s)
- Anja Schrade
- Children's Hospital (A.S., A.K., O.A., M.P., T.-P.A., M.H.), University of Helsinki, Helsinki 00014, Finland; Institute of Biomedicine (O.A.), University of Helsinki, Helsinki 00014, Finland; School of Pharmacy (M.H.), University of Eastern Finland, Kuopio 70211, Finland; Institute of Applied Biotechnology (S.F.), University of Applied Sciences Biberach, Biberach 88400, Germany; Metabolomics Unit (V.V.), Institute for Molecular Medicine Finland, University of Helsinki 00014, Helsinki, Finland; Departments of Physiology and Pediatrics (J.T.), University of Turku, Turku 20520, Finland; and Departments of Pediatrics (A.S., M.P., D.B.W., M.H.) and Developmental Biology (D.B.W.), Washington University in St. Louis, St. Louis, Missouri 63110
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bergeron F, Nadeau G, Viger RS. GATA4 knockdown in MA-10 Leydig cells identifies multiple target genes in the steroidogenic pathway. Reproduction 2014; 149:245-57. [PMID: 25504870 DOI: 10.1530/rep-14-0369] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
GATA4 is an essential transcription factor required for the initiation of genital ridge formation, for normal testicular and ovarian differentiation at the time of sex determination, and for male and female fertility in adulthood. In spite of its crucial roles, the genes and/or gene networks that are ultimately regulated by GATA4 in gonadal tissues remain to be fully understood. This is particularly true for the steroidogenic lineages such as Leydig cells of the testis where many in vitro (promoter) studies have provided good circumstantial evidence that GATA4 is a key regulator of Leydig cell gene expression and steroidogenesis, but formal proof is still lacking. We therefore performed a microarray screening analysis of MA-10 Leydig cells in which Gata4 expression was knocked down using an siRNA strategy. Analysis identified several GATA4-regulated pathways including cholesterol synthesis, cholesterol transport, and especially steroidogenesis. A decrease in GATA4 protein was associated with decreased expression of steroidogenic genes previously suspected to be GATA4 targets such as Cyp11a1 and Star. Gata4 knockdown also led to an important decrease in other novel steroidogenic targets including Srd5a1, Gsta3, Hsd3b1, and Hsd3b6, as well as genes known to participate in cholesterol metabolism such as Scarb1, Ldlr, Soat1, Scap, and Cyp51. Consistent with the decreased expression of these genes, a reduction in GATA4 protein compromised the ability of MA-10 cells to produce steroids both basally and under hormone stimulation. These data therefore provide strong evidence that GATA4 is an essential transcription factor that sits atop of the Leydig cell steroidogenic program.
Collapse
Affiliation(s)
- Francis Bergeron
- ReproductionMother and Child Health, Room T3-67, Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Centre de Recherche en Biologie de la Reproduction (CRBR), 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2Department of Obstetrics and GynecologyFaculty of Medicine, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Gabriel Nadeau
- ReproductionMother and Child Health, Room T3-67, Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Centre de Recherche en Biologie de la Reproduction (CRBR), 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2Department of Obstetrics and GynecologyFaculty of Medicine, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Robert S Viger
- ReproductionMother and Child Health, Room T3-67, Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Centre de Recherche en Biologie de la Reproduction (CRBR), 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2Department of Obstetrics and GynecologyFaculty of Medicine, Laval University, Quebec City, Quebec, Canada G1K 7P4 ReproductionMother and Child Health, Room T3-67, Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec, Centre de Recherche en Biologie de la Reproduction (CRBR), 2705 Laurier Boulevard, Quebec City, Quebec, Canada G1V 4G2Department of Obstetrics and GynecologyFaculty of Medicine, Laval University, Quebec City, Quebec, Canada G1K 7P4
| |
Collapse
|
25
|
Affiliation(s)
- Holly A LaVoie
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina
| |
Collapse
|