1
|
Charoensri S, Rege J, Lee C, Marko X, Sherk W, Sholinyan J, Rainey WE, Turcu AF. Human Gonads Do Not Contribute to the Circulating Pool of 11-Oxygenated Androgens. J Clin Endocrinol Metab 2024:dgae420. [PMID: 38885296 DOI: 10.1210/clinem/dgae420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/23/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
CONTEXT Androstenedione (A4) and testosterone (T) are produced by both the adrenal glands and the gonads. The adrenal enzyme 11β-hydroxylase (CYP11B1) executes the final step in cortisol synthesis; CYP11B1 also uses A4 and T as substrates, generating 11-hydroxyandrostenedione and 11-hydroxytestosterone, respectively. It has been suggested that CYP11B1 is expressed in the gonads, yet the circulating levels of all 11-oxygenated androgens (11-oxyandrogens) are similar in males and females of reproductive ages, despite enormous differences in T. OBJECTIVE To assess the gonadal contribution to the circulating pool of 11-oxyandrogens. METHODS We used liquid chromatography-tandem mass spectrometry to measure 13 steroids, including traditional and 11-oxyandrogens in: (I) paired gonadal and peripheral vein blood samples obtained during gonadal venograms from 11 patients (7 women), median age 37 (range 31-51 years); and (II) 17 women, median age 57 (range 41-81 years) before and after bilateral salpingo-oophorectomy (BSO). We also compared CYP11B1, 17α-hydroxylase/17,20-lyase (CYP17A1), and 3β-hydroxysteroid dehydrogenase type 2 (HSD3B2) mRNA expression in adrenal, ovarian, and testicular tissue. RESULTS A4, T, estradiol, estrone, progesterone, 17α- and 16α-hydroxyprogesterone were all higher in gonadal veins vs. periphery (p < 0.05 for all), while four 11-oxyandrogens were similar between matched gonadal and peripheral vein samples. Equally, in women who underwent BSO, A4 (median [interquartile range]: 59.7 [47.7-67.6] ng/dL vs. 32.7 [27.4-47.8] ng/dL, p < 0.001) and T (24.1 [16.4-32.3] vs.15.5 [13.7-19.0] ng/dL, p < 0.001) declined, while 11-oxyandrogens remained stable. Gonadal tissue displayed negligible CYP11B1 mRNA. CONCLUSION Despite producing substantial amounts of A4 and T, human gonads are not relevant sources of 11-oxyandrogens.
Collapse
Affiliation(s)
- Suranut Charoensri
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40000, Thailand
| | - Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Chaelin Lee
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Xhorlina Marko
- Division of Vascular and Interventional Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - William Sherk
- Division of Vascular and Interventional Radiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Julieta Sholinyan
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Adina F Turcu
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
2
|
You Z, Yuan J, Wang Y, Sun Y, Ni A, Li Y, Ma H, Ma T, Chen J. Integrated transcriptomic analysis on chicken ovary reveals CYP21A1 affects follicle granulosa cell development and steroid hormone synthesis. Poult Sci 2024; 103:103589. [PMID: 38471223 PMCID: PMC11067781 DOI: 10.1016/j.psj.2024.103589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Egg production is an economically important trait in poultry breeding and production. Follicular development was regulated by several hormones released and genes expressed in the granulosa cells, impacting the egg production and fecundity of hens. However, the molecular functions of these candidate genes that modulate these processes remain largely unknown. In the present study, bioinformatics analyses were performed to identify the candidate genes related to egg production in the ovarian tissue of White Leghorns with high egg production and Beijing You chicken with low egg production during sexual maturity and peak laying periods. The ovarian granulosa cells were used to assess the function of CYP21A1 by transfecting with CYP21A1-specific small interfering RNAs (siRNAs) and overexpression plasmids. We identified 514 differentially expressed genes (|Log2(fold change) | >1, P <0.05) between the 2 chicken breeds in both laying periods. Among these genes, CYP21A1, which is involved in the steroid hormone biosynthesis pathway was consistently upregulated in White Leghorns. Weighted gene co-expression network analysis (WGCNA) further suggested that CYP21A1 was a hub gene, which could positively respond to treatment with follicle stimulation hormone (FSH), affecting egg production. The interference of CYP21A1 significantly inhibited cell proliferation and promoted cell apoptosis. Overexpression of CYP21A1 promotes cell proliferation and inhibits cell apoptosis. Furthermore, the interference with CYP21A1 significantly downregulated the expression of STAR, CYP11A1, HSD3B1, and FSHR and also decreased the synthesis of progesterone (P4) and estradiol (E2) in granulosa cells. Overexpression of CYP21A1 increased the synthesis of P4 and estradiol E2 and the expression of steroid hormone synthesis-related genes in granulosa cells. Our findings provide new evidence for the biological role of CYP21A1 on granulosa cell proliferation, apoptosis, and steroid hormone synthesis, which lays the theoretical basis for improving egg production.
Collapse
Affiliation(s)
- Zhangjing You
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, 056038, Hebei, China
| | - Jingwei Yuan
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yuanmei Wang
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018 China
| | - Yanyan Sun
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Aixin Ni
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yunlei Li
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Hui Ma
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Tenghe Ma
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, 056038, Hebei, China
| | - Jilan Chen
- State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
3
|
Weniger M, Mattes M, Grünewald TGP, Köhler K, Hübner A, Beuschlein F, Reisch N. Quantitative Characterization of Ectopic Adrenal Gene Expression in Fetal Testes in 21-Hydroxylase Deficient Mice. Horm Metab Res 2024; 56:38-44. [PMID: 38171371 DOI: 10.1055/a-2216-0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Testicular adrenal rest tumors (TART) are a frequent and fertility impairing long-term complication in males with classic congenital adrenal hyperplasia. Due to lack of clear experimental data on their origin, they are hypothesized to be derived from ectopic adrenocortical cells within testicular tissue mainly growing upon stimulation by chronically elevated levels of adrenocorticotropin (ACTH). Alternatively, a more totipotent embryological origin has been discussed as the potential source of these tumors. The aim of this study was to quantify alterations of ectopic expression of adrenocortical genes (CYP11B1, CYP11B2, CYP21, MC2R) and the Leydig cell specific marker (INSL3) in testicular tissue of fetal 21-hydroxylase deficient (21OHD) mice. Timed-pregnancy studies were performed using H-2aw18 (aw18)-mice. Testes and adrenals of E15.5 and E18.5 mouse fetuses were used for real-time PCR and immunohistochemistry. Gene expression levels were analyzed for genotype-dependent alterations and compared with immunohistochemistry. While enzymes of steroidogenesis showed a significant increased expression in adrenals of 21OHD mice at both E15.5 and E18.5 compared to wild-type (WT) mice, expression levels were unaltered in testes of 21OHD mice. When compared to WT adrenals a significant increase of INSL3 expression in adrenals of 21OHD mice at E15.5 and E18.5 was detected. Cells with adrenocortical properties in mice fetal testis differ from in situ adrenocortical cells in gene expression and growth at E15.5 and E18.5. These findings suggest that the different local regulation and different local niche in adrenals and testes influence growth of aberrant adrenal cells.
Collapse
Affiliation(s)
| | - Maria Mattes
- Medizinische Klinik IV, LMU Klinikum München, Munich, Germany
| | - Thomas G P Grünewald
- Max Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, LMU Klinikum München, Munich, Germany
| | - Katrin Köhler
- Pediatric Endocrinology, Children's Hospital, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Angela Hübner
- Pediatric Endocrinology, Children's Hospital, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Felix Beuschlein
- Medizinische Klinik IV, LMU Klinikum München, Munich, Germany
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, UniversitätsSpital Zürich, Zürich, Switzerland
| | - Nicole Reisch
- Medizinische Klinik IV, LMU Klinikum München, Munich, Germany
| |
Collapse
|
4
|
Jin J, Li K, Du Y, Gao F, Wang Z, Li W. Multi-omics study identifies that PICK1 deficiency causes male infertility by inhibiting vesicle trafficking in Sertoli cells. Reprod Biol Endocrinol 2023; 21:114. [PMID: 38001535 PMCID: PMC10675906 DOI: 10.1186/s12958-023-01163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Infertility affects approximately 10-15% of reproductive-age men worldwide, and genetic causes play a role in one-third of cases. As a Bin-Amphiphysin-Rvs (BAR) domain protein, protein interacting with C-kinase 1 (PICK1) deficiency could lead to impairment of acrosome maturation. However, its effects on auxiliary germ cells such as Sertoli cells are unknown. PURPOSE The present work was aimed to use multi-omics analysis to research the effects of PICK1 deficiency on Sertoli cells and to identify effective biomarkers to distinguish fertile males from infertile males caused by PICK1 deficiency. METHODS Whole-exome sequencing (WES) was performed on 20 infertility patients with oligozoospermia to identify pathogenic PICK1 mutations. Multi-omics analysis of a PICK1 knockout (KO) mouse model was utilized to identify pathogenic mechanism. Animal and cell function experiments of Sertoli cell-specific PICK1 KO mouse were performed to verify the functional impairment of Sertoli cells. RESULTS Two loss-of-function deletion mutations c.358delA and c.364delA in PICK1 resulting in transcription loss of BAR functional domain were identified in infertility patients with a specific decrease in serum inhibin B, indicating functional impairment of Sertoli cells. Multi-omics analysis of PICK1 KO mouse illustrated that targeted genes of differentially expressed microRNAs and mRNAs are significantly enriched in the negative regulatory role in the vesicle trafficking pathway, while metabolomics analysis showed that the metabolism of amino acids, lipids, cofactors, vitamins, and endocrine factors changed. The phenotype of PICK1 KO mouse showed a reduction in testis volume, a decreased number of mature spermatozoa and impaired secretory function of Sertoli cells. In vitro experiments confirmed that the expression of growth factors secreted by Sertoli cells in PICK1 conditional KO mouse such as Bone morphogenetic protein 4 (BMP4) and Fibroblast growth factor 2 (FGF2) were decreased. CONCLUSIONS Our study attributed male infertility caused by PICK1 deficiency to impaired vesicle-related secretory function of Sertoli cells and identified a variety of significant candidate biomarkers for male infertility induced by PICK1 deficiency.
Collapse
Affiliation(s)
- Jing Jin
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratories, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Kaiqiang Li
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Yaoqiang Du
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Fang Gao
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Zhen Wang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China.
| | - Weixing Li
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratories, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
5
|
Schröder MAM, Greenald D, Lodewijk R, van Herwaarden AE, Span PN, Sweep FCGJ, Mitchell RT, Claahsen-van der Grinten HL. Evaluation of Ex Vivo Adrenocorticotropic Hormone Responsiveness of Human Fetal Testis. Endocrinology 2023; 164:bqad165. [PMID: 37935047 PMCID: PMC10652325 DOI: 10.1210/endocr/bqad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
Testicular adrenal rest tumors (TARTs), commonly occurring in males with congenital adrenal hyperplasia, may arise from chronic stimulation of adrenocorticotropic hormone (ACTH)-sensitive cells in the testes. It is not yet established whether the human fetal testis (HFT) is responsive to ACTH. To investigate this, we cultured HFT tissue with and without ACTH for up to 5 days, and quantified adrenal steroid hormones and expression of adrenal steroidogenic enzymes. Fetal testis and adrenal tissue produced high levels of testosterone and cortisol, respectively, indicating viability. In contrast to fetal adrenal tissues, the expression of ACTH receptor MC2R was either absent or expressed at extremely low levels in ex vivo HFT tissue and no clear response to ACTH in gene expression or steroid hormone production was observed. Altogether, this study suggests that the HFT is unresponsive to ACTH, which would indicate that a TART does not arise from fetal testicular cells chronically exposed to ACTH in utero.
Collapse
Affiliation(s)
- Mariska A M Schröder
- Department of Pediatrics, Radboud Amalia Children's Hospital, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboudumc Graduate School, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
- MRC Centre for Reproductive Health, Institute for Regeneration and Repair, The University of Edinburgh, and the Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, UK
| | - David Greenald
- MRC Centre for Reproductive Health, Institute for Regeneration and Repair, The University of Edinburgh, and the Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, UK
| | - Renate Lodewijk
- Department of Laboratory Medicine, Radboudumc Graduate School, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Antonius E van Herwaarden
- Department of Laboratory Medicine, Radboudumc Graduate School, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Paul N Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboudumc Graduate School, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboudumc Graduate School, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, Institute for Regeneration and Repair, The University of Edinburgh, and the Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, UK
| | - Hedi L Claahsen-van der Grinten
- Department of Pediatrics, Radboud Amalia Children's Hospital, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
6
|
Fanelli F, Magagnoli M, Mezzullo M, Lispi M, Limoncella S, Tommasini A, Pelusi C, Santi D, Simoni M, Pagotto U, Casarini L. Exploring the human chorionic gonadotropin induced steroid secretion profile of mouse Leydig tumor cell line 1 by a 20 steroid LC-MS/MS panel. J Steroid Biochem Mol Biol 2023; 229:106270. [PMID: 36764496 DOI: 10.1016/j.jsbmb.2023.106270] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/29/2022] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The canonical androgen synthesis in Leydig cells involves Δ5 and Δ4 steroids. Besides, the backdoor pathway, eompassing 5α and 5α,3α steroids, is gaining interest in fetal and adult pathophysiology. Moreover, the role of androgen epimers and progesterone metabolites is still unknown. We developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for measuring 20 steroids and used it to investigate the steroid secretion induced by human chorionic gonadotropin (hCG) in the mouse Leydig tumor cell line 1 (mLTC1). Steroids were extracted from 500 µL supernatants from unstimulated or 100 pM hCG-exposed mLTC1 cells, separated on a Luna C8 100 × 3 mm, 3 µm column, with 100 µM NH4F and methanol as mobile phases, and analyzed by positive electrospray ionization and multiple reaction monitoring. Sensitivity ranged within 0.012-38.0 nmol/L. Intra-assay and inter-assay imprecision were < 9.1% and 10.0%, respectively. Trueness, recovery and matrix factor were within 93.4-122.0, 55.6-104.1 and 76.4-106.3%, respectively. Levels of 16OH-progesterone, 11-deoxycortisol, androstenedione, 11-deoxycorticosterone, testosterone, 17OH-progesterone, androstenedione, epitestosterone, dihydrotestosterone, progesterone, androsterone and 17OH-allopregnanolone were effectively measured. Traces of 17OH-dihydroprogesterone, androstanediol and dihydroprogesterone were found, whereas androstenediol, 17OH-pregnenolone, dehydroepiandrosterone, pregnenolone and allopregnanolone showed no peak. hCG induced an increase of 80.2-102.5 folds in 16OH-progesterone, androstenedione and testosterone, 16.6 in dihydrotestosterone, 12.2-27.5 in epitestosterone, progesterone and metabolites, 8.1 in 17OH-allopregnanolone and ≤ 3.3 in 5α and 5α,3α steroids. In conclusion, our LC-MS/MS method allows exploring the Leydig steroidogenesis flow according to multiple pathways. Beside the expected stimulation of the canonical pathway, hCG increased progesterone metabolism and, to a low extent, the backdoor route.
Collapse
Affiliation(s)
- Flaminia Fanelli
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy.
| | - Matteo Magagnoli
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy
| | - Marco Mezzullo
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy
| | - Monica Lispi
- International Ph.D. School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy; Global Medical Affair, Merck KGaA, Darmstadt, Germany
| | - Silvia Limoncella
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy
| | - Alessia Tommasini
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy
| | - Carla Pelusi
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy; Endocrinology and Prevention and Care of Diabetes Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Uberto Pagotto
- Endocrinology research group, Center for Applied Biomedical Research, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Italy; Endocrinology and Prevention and Care of Diabetes Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia. Ospedale di Baggiovara, Via P. Giardini 1355, 41126 Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| |
Collapse
|
7
|
Gray LE, Lambright CS, Conley JM, Evans N, Furr JR, Hannas BR, Wilson VS, Sampson H, Foster PMD. Genomic and Hormonal Biomarkers of Phthalate-Induced Male Rat Reproductive Developmental Toxicity Part II: A Targeted RT-qPCR Array Approach That Defines a Unique Adverse Outcome Pathway. Toxicol Sci 2021; 182:195-214. [PMID: 33983380 DOI: 10.1093/toxsci/kfab053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previously, we demonstrated that exposure to some diortho-phthalate esters during sexual differentiation disrupts male reproductive development by reducing fetal rat testis testosterone production (T Prod) and gene expression in a dose-related manner. The objectives of the current project were to expand the number of test compounds that might reduce fetal T Prod, including phthalates, phthalate alternatives, pesticides, and drugs, and to compare reductions in T Prod with altered testis mRNA expression. We found that PEs that disrupt T Prod also reduced expression of a unique "cluster" of mRNAs for about 35 genes related to sterol transport, testosterone and insulin-like hormone 3 hormone syntheses, and lipoprotein signaling and cholesterol synthesis. However, phthalates had little or no effect on mRNA expression of genes in peroxisome proliferator-activated receptor (PPAR) pathways in the fetal liver, whereas the 3 PPAR agonists induced the expression of mRNA for multiple fetal liver PPAR pathway genes without reducing testis T Prod. In summary, phthalates that disrupt T Prod act via a novel adverse outcome pathway including down regulation of mRNA for genes involved in fetal endocrine function and cholesterol synthesis and metabolism. This profile was not displayed by PEs that did not reduce T Prod, PPAR agonists or the other chemicals. Reductions in fetal testis gene expression and T Prod in utero can be used to establish relative potency factors that can be used quantitatively to predict the doses of individual PEs and mixtures of phthalates that produce adverse reproductive tract effects in male offspring.
Collapse
Affiliation(s)
- Leon Earl Gray
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | - Christy S Lambright
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | - Justin M Conley
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | - Nicola Evans
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | | | - Bethany R Hannas
- Corteva, Agriscience, Haskell R&D Center, Newark, Delaware 19711, USA
| | - Vickie S Wilson
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | - Hunter Sampson
- Reproductive and Developmental Toxicology Branch, PHITD, CPHEA, ORD, US Environmental Protection Agency, North Carolina 27711, USA
| | | |
Collapse
|
8
|
Han S, Baba T, Yanai S, Byun DJ, Morohashi KI, Kim JH, Choi MH. GC-MS-based metabolic signatures reveal comparative steroidogenic pathways between fetal and adult mouse testes. Andrology 2020; 9:400-406. [PMID: 32810374 DOI: 10.1111/andr.12893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/17/2020] [Accepted: 08/14/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND Previous studies on gonadal steroidogenesis have not compared metabolic pathways between fetal and adult mouse testes to date. OBJECTIVES To evaluate comparative metabolic signatures of testicular steroids between fetus and adult mice using gas chromatography-mass spectrometry (GC-MS)-based steroid profiling. MATERIALS AND METHODS GC-MS with molecular-specific scan modes was optimized for selective and sensitive detection of 23 androgens, 7 estrogens, 14 progestogens, and 13 corticoids from mouse testes with a quantification limit of 0.1-5.0 ng/mL and reproducibility (coefficient of variation: 0.3%-19.9%). Based on 26 steroids quantitatively detected in testes, comparative steroid signatures were analyzed for mouse testes of 8 fetuses on embryonic day 16.5 and 8 adults on postnatal days 56-60. RESULTS In contrast to large amounts of steroids in adult testes (P < .0002), all testicular levels per weight unit of protein were significantly increased in fetal testes (P < .002, except 6β-hydroxytestosterone of P = .065). Both 11β-hydroxyandrostenedione and 7α-hydroxytestosterone were only measurable in fetal testes, and metabolic ratios of testosterone to androstenediol and androstenedione were also increased in fetal testes (P < .05 for both). DISCUSSION AND CONCLUSION Testicular steroid signatures showed that both steroidogenic Δ4 and Δ5 pathways in the production of testosterone were activated more during prenatal development. Both 7α- and 11β-hydroxylations were predominant, while hydroxylations at C-6, C-15, and C-16 of testosterone and androstenedione were decreased in the fetus. The present GC-MS-based steroid profiling may facilitate understanding of the development of testicular steroidogenesis.
Collapse
Affiliation(s)
- Soyun Han
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea.,College of Life Sciences, Korea University, Seoul, Korea
| | - Takashi Baba
- Department of Molecular Biology, Kyushu University, Fukuoka, Japan
| | - Shogo Yanai
- Department of Molecular Biology, Kyushu University, Fukuoka, Japan
| | - Dong Jun Byun
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea
| | | | - Jae-Hong Kim
- College of Life Sciences, Korea University, Seoul, Korea
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Korea
| |
Collapse
|
9
|
Grinspon RP, Rey RA. Molecular Characterization of XX Maleness. Int J Mol Sci 2019; 20:ijms20236089. [PMID: 31816857 PMCID: PMC6928850 DOI: 10.3390/ijms20236089] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
Androgens and anti-Müllerian hormone (AMH), secreted by the foetal testis, are responsible for the development of male reproductive organs and the regression of female anlagen. Virilization of the reproductive tract in association with the absence of Müllerian derivatives in the XX foetus implies the existence of testicular tissue, which can occur in the presence or absence of SRY. Recent advancement in the knowledge of the opposing gene cascades driving to the differentiation of the gonadal ridge into testes or ovaries during early foetal development has provided insight into the molecular explanation of XX maleness.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
- Correspondence: (R.P.G.); (R.A.R.); Tel.: +54-11-49635931 (R.P.G.)
| |
Collapse
|
10
|
O'Shaughnessy PJ, Mitchell RT, Monteiro A, O'Hara L, Cruickshanks L, der Grinten HCV, Brown P, Abel M, Smith LB. Androgen receptor expression is required to ensure development of adult Leydig cells and to prevent development of steroidogenic cells with adrenal characteristics in the mouse testis. BMC DEVELOPMENTAL BIOLOGY 2019; 19:8. [PMID: 30995907 PMCID: PMC6472051 DOI: 10.1186/s12861-019-0189-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/29/2019] [Indexed: 01/10/2023]
Abstract
Background The interstitium of the mouse testis contains Leydig cells and a small number of steroidogenic cells with adrenal characteristics which may be derived from the fetal adrenal during development or may be a normal subset of the developing fetal Leydig cells. Currently it is not known what regulates development and/or proliferation of this sub-population of steroidogenic cells in the mouse testis. Androgen receptors (AR) are essential for normal testicular function and in this study we have examined the role of the AR in regulating interstitial cell development. Results Using a mouse model which lacks gonadotropins and AR (hpg.ARKO), stimulation of luteinising hormone receptors in vivo with human chorionic gonadotropin (hCG) caused a marked increase in adrenal cell transcripts/protein in a group of testicular interstitial cells. hCG also induced testicular transcripts associated with basic steroidogenic function in these mice but had no effect on adult Leydig cell-specific transcript levels. In hpg mice with functional AR, treatment with hCG induced Leydig cell-specific function and had no effect on adrenal transcript levels. Examination of mice with cell-specific AR deletion and knockdown of AR in a mouse Leydig cell line suggests that AR in the Leydig cells are likely to regulate these effects. Conclusions This study shows that in the mouse the androgen receptor is required both to prevent development of testicular cells with adrenal characteristics and to ensure development of an adult Leydig cell phenotype. Electronic supplementary material The online version of this article (10.1186/s12861-019-0189-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter J O'Shaughnessy
- College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, G61 1QH, Glasgow, UK.
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Ana Monteiro
- College of Medical, Veterinary and Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, G61 1QH, Glasgow, UK
| | - Laura O'Hara
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Lyndsey Cruickshanks
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Hedi Claahsen-van der Grinten
- Department of Paediatrics, Radboud Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pamela Brown
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Margaret Abel
- Department of Human Anatomy and Genetics, University of Oxford, South Parks Rd, Oxford, OX1 3QX, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| |
Collapse
|
11
|
Atorvastatin decreases steroid production in H295R cells and in major endocrine tissues of male rats. Arch Toxicol 2018; 92:1703-1715. [DOI: 10.1007/s00204-018-2187-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/13/2018] [Indexed: 12/13/2022]
|
12
|
Munkboel CH, Larsen LW, Weisser JJ, Møbjerg Kristensen D, Styrishave B. Sertraline Suppresses Testis and Adrenal Steroid Production and Steroidogenic Gene Expression While Increasing LH in Plasma of Male Rats Resulting in Compensatory Hypogonadism. Toxicol Sci 2018; 163:609-619. [DOI: 10.1093/toxsci/kfy059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Cecilie Hurup Munkboel
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - Lizette Weber Larsen
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - Johan Juhl Weisser
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - David Møbjerg Kristensen
- Danish Headache Center, Department of Neurology, Rigshospitalet, University of Copenhagen, 1165 Copenhagen, Denmark
- Inserm (Institut National de la Santé et de la Recherche Médicale), Irset – Inserm UMR 1085, 35000 Rennes, France
| | - Bjarne Styrishave
- Toxicology Laboratory, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| |
Collapse
|
13
|
Imamichi Y, Yuhki KI, Orisaka M, Kitano T, Mukai K, Ushikubi F, Taniguchi T, Umezawa A, Miyamoto K, Yazawa T. 11-Ketotestosterone Is a Major Androgen Produced in Human Gonads. J Clin Endocrinol Metab 2016; 101:3582-3591. [PMID: 27428878 DOI: 10.1210/jc.2016-2311] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CONTEXT 11-ketotestosterone (11-KT) is a novel class of active androgen. However, the detail of its synthesis remains unknown for humans. OBJECTIVE The objective of this study was to clarify the production and properties of 11-KT in human. Design, Participants, and Methods: Expression of cytochrome P450 and 11β-hydroxysteroid dehydrogenase types 1 and 2 (key enzymes involved in the synthesis of 11-KT) were investigated in human gonads. The production of 11-KT was investigated in Leydig cells. Plasma concentrations of testosterone and 11-KT were measured in 10 women and 10 men of reproductive age. Investigation of its properties was performed using breast cancer-derived MCF-7 cells. RESULTS Cytochrome P450 and 11β-hydroxysteroid dehydrogenase types 1 and 2 were detected in Leydig cells and theca cells. Leydig cells produced 11-KT, and relatively high levels of plasma 11-KT were measured in both men and women. There was no sexual dimorphism in the plasma levels of 11-KT, even though testosterone levels were more than 20 times higher in men than in women. It is noteworthy that the levels of testosterone and 11-KT were similar in women. In a luciferase reporter system, 11-KT activated human androgen receptor-mediated transactivation. Conversely, 11-KT did not activate estrogen receptor-mediated transactivation in aromatase-expressed MCF-7 cells, whereas testosterone did following conversion to estrogen. 11-KT did not affect the estrogen/estrogen receptor -mediated cell proliferation of MCF-7 cells. Furthermore, it significantly inhibited cell proliferation when androgen receptor was transfected into MCF-7 cells. CONCLUSIONS The current study indicates that 11-KT is produced in the gonads and represents a major androgen in human. It can potentially serve as a nonaromatizable androgen.
Collapse
Affiliation(s)
- Yoshitaka Imamichi
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Koh-Ichi Yuhki
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Makoto Orisaka
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Takeshi Kitano
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Kuniaki Mukai
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Fumitaka Ushikubi
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Takanobu Taniguchi
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Akihiro Umezawa
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Kaoru Miyamoto
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Takashi Yazawa
- Departments of Pharmacology (Y.I., K.-i.Y., F.U.) and Biochemistry (T.T., T.Y.), Asahikawa Medical University, Hokkaido 078-8510, Japan; Departments of Biochemistry (Y.I., K.Mi.) and Obstetrics and Gynecology (M.O.), Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Department of Materials and Life Science (T.K.), Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan; Department of Biochemistry and Medical Education Center (K.Mu.), Keio University School of Medicine, Tokyo 160-8582, Japan; and Department of Reproduction (A.U.), National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
14
|
Tevosian SG, Jiménez E, Hatch HM, Jiang T, Morse DA, Fox SC, Padua MB. Adrenal Development in Mice Requires GATA4 and GATA6 Transcription Factors. Endocrinology 2015; 156:2503-17. [PMID: 25933105 PMCID: PMC4475720 DOI: 10.1210/en.2014-1815] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The adrenal glands consist of an outer cortex and an inner medulla, and their primary purposes include hormone synthesis and secretion. The adrenal cortex produces a complex array of steroid hormones, whereas the medulla is part of the sympathetic nervous system and produces the catecholamines epinephrine and norepinephrine. In the mouse, GATA binding protein (GATA) 4 and GATA6 transcription factors are coexpressed in several embryonic tissues, including the adrenal cortex. To explore the roles of GATA4 and GATA6 in mouse adrenal development, we conditionally deleted these genes in adrenocortical cells using the Sf1Cre strain of animals. We report here that mice with Sf1Cre-mediated double deletion of Gata4 and Gata6 genes lack identifiable adrenal glands, steroidogenic factor 1-positive cortical cells and steroidogenic gene expression in the adrenal location. The inactivation of the Gata6 gene alone (Sf1Cre;Gata6(flox/flox)) drastically reduced the adrenal size and corticosterone production in the adult animals. Adrenocortical aplasia is expected to result in the demise of the animal within 2 weeks after birth unless glucocorticoids are provided. In accordance, Sf1Cre;Gata4(flox/flox)Gata6(flox/flox) females depend on steroid supplementation to survive after weaning. Surprisingly, Sf1Cre;Gata4(flox/flox)Gata6(flox/flox) males appear to live normal lifespans as vital steroidogenic synthesis shifts to their testes. Our results reveal a requirement for GATA factors in adrenal development and provide a novel tool to characterize the transcriptional network controlling adrenocortical cell fates.
Collapse
Affiliation(s)
- Sergei G Tevosian
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Elizabeth Jiménez
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Heather M Hatch
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Tianyu Jiang
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Deborah A Morse
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Shawna C Fox
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| | - Maria B Padua
- Department of Physiological Sciences (S.G.T., E.J., H.M.H., T.J., S.C.F., M.B.P.), College of Veterinary Medicine, University of Florida, Gainesville, Florida 32610-0144; and Department of Applied Physiology and Kinesiology (D.A.M.), College of Health and Human Performance, University of Florida, Gainesville, Florida 32611-8200
| |
Collapse
|
15
|
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: 0.7] [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
|
16
|
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: 4.2] [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
|
17
|
Schillebeeckx M, Pihlajoki M, Gretzinger E, Yang W, Thol F, Hiller T, Löbs AK, Röhrig T, Schrade A, Cochran R, Jay PY, Heikinheimo M, Mitra RD, Wilson DB. Novel markers of gonadectomy-induced adrenocortical neoplasia in the mouse and ferret. Mol Cell Endocrinol 2015; 399:122-30. [PMID: 25289806 PMCID: PMC4262703 DOI: 10.1016/j.mce.2014.09.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 07/18/2014] [Accepted: 09/29/2014] [Indexed: 12/25/2022]
Abstract
Gonadectomy (GDX) induces sex steroid-producing adrenocortical tumors in certain mouse strains and in the domestic ferret. Transcriptome analysis and DNA methylation mapping were used to identify novel genetic and epigenetic markers of GDX-induced adrenocortical neoplasia in female DBA/2J mice. Markers were validated using a combination of laser capture microdissection, quantitative RT-PCR, in situ hybridization, and immunohistochemistry. Microarray expression profiling of whole adrenal mRNA from ovariectomized vs. intact mice demonstrated selective upregulation of gonadal-like genes including Spinlw1 and Insl3 in GDX-induced adrenocortical tumors of the mouse. A complementary candidate gene approach identified Foxl2 as another gonadal-like marker expressed in GDX-induced neoplasms of the mouse and ferret. That both "male-specific" (Spinlw1) and "female-specific" (Foxl2) markers were identified is noteworthy and implies that the neoplasms exhibit mixed characteristics of male and female gonadal somatic cells. Genome-wide methylation analysis showed that two genes with hypomethylated promoters, Igfbp6 and Foxs1, are upregulated in GDX-induced adrenocortical neoplasms. These new genetic and epigenetic markers may prove useful for studies of steroidogenic cell development and for diagnostic testing.
Collapse
Affiliation(s)
- Maximiliaan Schillebeeckx
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - 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
| | - Elisabeth Gretzinger
- 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
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Franziska Thol
- 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
| | - Theresa Hiller
- 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
| | - Ann-Kathrin Löbs
- 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
| | - 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
| | - 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
| | - Rebecca Cochran
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Patrick Y Jay
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Pediatrics, 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
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - 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
|
18
|
Rebourcet D, O'Shaughnessy PJ, Pitetti JL, Monteiro A, O'Hara L, Milne L, Tsai YT, Cruickshanks L, Riethmacher D, Guillou F, Mitchell RT, van't Hof R, Freeman TC, Nef S, Smith LB. Sertoli cells control peritubular myoid cell fate and support adult Leydig cell development in the prepubertal testis. Development 2014; 141:2139-49. [PMID: 24803659 DOI: 10.1242/dev.107029] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sertoli cells (SCs) regulate testicular fate in the differentiating gonad and are the main regulators of spermatogenesis in the adult testis; however, their role during the intervening period of testis development, in particular during adult Leydig cell (ALC) differentiation and function, remains largely unknown. To examine SC function during fetal and prepubertal development we generated two transgenic mouse models that permit controlled, cell-specific ablation of SCs in pre- and postnatal life. Results show that SCs are required: (1) to maintain the differentiated phenotype of peritubular myoid cells (PTMCs) in prepubertal life; (2) to maintain the ALC progenitor population in the postnatal testis; and (3) for development of normal ALC numbers. Furthermore, our data show that fetal LCs function independently from SC, germ cell or PTMC support in the prepubertal testis. Together, these findings reveal that SCs remain essential regulators of testis development long after the period of sex determination. These findings have significant implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.
Collapse
Affiliation(s)
- Diane Rebourcet
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Castellanos CG, Sørvik IB, Tanum MB, Verhaegen S, Brandt I, Ropstad E. Differential effects of the persistent DDT metabolite methylsulfonyl-DDE in nonstimulated and LH-stimulated neonatal porcine Leydig cells. Toxicol Appl Pharmacol 2013; 267:247-55. [DOI: 10.1016/j.taap.2012.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 12/07/2012] [Accepted: 12/28/2012] [Indexed: 11/25/2022]
|
20
|
Hannas BR, Lambright CS, Furr J, Evans N, Foster PMD, Gray EL, Wilson VS. Genomic biomarkers of phthalate-induced male reproductive developmental toxicity: a targeted RT-PCR array approach for defining relative potency. Toxicol Sci 2011; 125:544-57. [PMID: 22112501 DOI: 10.1093/toxsci/kfr315] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Male rat fetuses exposed to certain phthalate esters (PEs) during sexual differentiation display reproductive tract malformations due to reductions in testosterone (T) production and the expression of steroidogenesis- and INSL3-related genes. In the current study, we used a 96-well real-time PCR array containing key target genes representing sexual determination and differentiation, steroidogenesis, gubernaculum development, and androgen signaling pathways to rank the relative potency of several PEs. We executed dose-response studies with diisobutyl (DIBP), dipentyl (DPeP), dihexyl (DHP), diheptyl (DHeP), diisononyl (DINP), or diisodecyl phthalate (DIDP) and serial dilutions of a mixture of nine phthalates. All phthalates, with the exception of DIDP, reduced fetal testicular T production. Several genes involved in cholesterol transport, androgen synthesis, and Insl3 also were downregulated in a dose-responsive manner by DIBP, DPeP, DHP, DHeP, DINP, and the 9-PE mixture. Despite speculation of peroxisome proliferator activated receptor (PPAR) involvement in the effects of PEs on the fetal testis, no PPAR-related genes were affected in the fetal testes by exposure to any of the tested PEs. Furthermore, the potent PPARα agonist, Wy-14,643, did not reduce fetal testicular T production following gestational day 14-18 exposure, suggesting that the antiandrogenic activity of PEs is not PPARα mediated. The overall sensitivity of the fetal endpoints (gene expression or T production) for the six phthalates from most to least was Cyp11b1 > Star = Scarb1 > Cyp17a1 = T production > Cyp11a1 = Hsd3b = Insl3 > Cyp11b2. The overall potency of the individual phthalates was DPeP > DHP > DIBP ≥ DHeP > DINP. Finally, the observed mixture interaction was adequately modeled by the dose-addition model for most of the affected genes. Together, these data advance our understanding of the collective reproductive toxicity of the PE compounds.
Collapse
Affiliation(s)
- Bethany R Hannas
- Reproductive Toxicology Branch, Toxicology Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Lardenois A, Chalmel F, Barrionuevo F, Demougin P, Scherer G, Primig M. Profiling spermatogenic failure in adult testes bearing Sox9-deficient Sertoli cells identifies genes involved in feminization, inflammation and stress. Reprod Biol Endocrinol 2010; 8:154. [PMID: 21182756 PMCID: PMC3024295 DOI: 10.1186/1477-7827-8-154] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 12/23/2010] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Sox9 (Sry box containing gene 9) is a DNA-binding transcription factor involved in chondrocyte development and sex determination. The protein's absence in testicular Sertoli nurse cells has been shown to disrupt testicular function in adults but little is known at the genome-wide level about molecular events concomitant with testicular break-down. METHODS To determine the genome-wide effect on mRNA concentrations triggered by the absence of Sox9 in Sertoli cells we analysed adult testicular tissue from wild-type versus mutant mice with high-density oligonucleotide microarrays and integrated the output of this experiment with regulatory motif predictions and protein-protein network data. RESULTS We report the genome-wide mRNA signature of adult testes lacking Sox9 in Sertoli cells before and after the onset of late spermatogenic failure as compared to fertile controls. The GeneChip data integrated with evolutionarily conserved Sox9 DNA binding motifs and regulatory network data identified genes involved in feminization, stress response and inflammation. CONCLUSIONS Our results extend previous observations that genes required for female gonadogenesis are up-regulated in the absence of Sox9 in fetal Sertoli cells to the adult stage. Importantly, we identify gene networks involved in immunological processes and stress response which is reminiscent of a phenomenon occurring in a sub-group of infertile men. This suggests mice lacking Sox9 in their Sertoli cells to be a potentially useful model for adult human testicular failure.
Collapse
Affiliation(s)
- Aurélie Lardenois
- Inserm, U625, Université de Rennes 1, IFR140, Rennes, F-35042, France
| | | | | | | | | | | |
Collapse
|
22
|
Val P, Swain A. Gene dosage effects and transcriptional regulation of early mammalian adrenal cortex development. Mol Cell Endocrinol 2010; 323:105-14. [PMID: 20025938 DOI: 10.1016/j.mce.2009.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Pierre Val
- Centre National de la Recherche Scientifique, Unité mixte de Recherche 6247, Génétique, Reproduction et Développement, Clermont Université, 63177 Aubière, France
| | | |
Collapse
|
23
|
Lavoie HA, King SR. Transcriptional regulation of steroidogenic genes: STARD1, CYP11A1 and HSD3B. Exp Biol Med (Maywood) 2009; 234:880-907. [PMID: 19491374 DOI: 10.3181/0903-mr-97] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Expression of the genes that mediate the first steps in steroidogenesis, the steroidogenic acute regulatory protein (STARD1), the cholesterol side-chain cleavage enzyme, cytochrome P450scc (CYP11A1) and 3beta-hydroxysteroid dehydrogenase/Delta5-Delta4 isomerase (HSD3B), is tightly controlled by a battery of transcription factors in the adrenal cortex, the gonads and the placenta. These genes generally respond to the same hormones that stimulate steroid production through common pathways such as cAMP signaling and common actions on their promoters by proteins such as NR5A and GATA family members. However, there are distinct temporal, tissue and species-specific differences in expression between the genes that are defined by combinatorial regulation and unique promoter elements. This review will provide an overview of the hormonal and transcriptional regulation of the STARD1, CYP11A1 and specific steroidogenic HSD3B genes in the adrenal, testis, ovary and placenta and discuss the current knowledge regarding the key transcriptional factors involved.
Collapse
Affiliation(s)
- Holly A Lavoie
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| | | |
Collapse
|
24
|
Griswold SL, Behringer RR. Fetal Leydig cell origin and development. Sex Dev 2009; 3:1-15. [PMID: 19339813 DOI: 10.1159/000200077] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 10/29/2008] [Indexed: 11/19/2022] Open
Abstract
Male sexual differentiation is a complex process requiring the hormone-producing function of somatic cells in the gonad, including Sertoli cells and fetal Leydig cells (FLCs). FLCs are essential for virilization of the male embryo, but despite their crucial function, relatively little is known about their origins or development. Adult Leydig cells (ALCs), which arise at puberty, have been studied extensively and much of what has been learned about this cell population has been extrapolated to FLCs. This approach is problematic in that prevailing dogma in the field asserts that these 2 populations are distinct in origin. As such, it is imprudent to assume that FLCs arise and develop in a similar manner to ALCs. This review provides a critical assessment of studies performed on FLC populations, rather than those extrapolated from ALC studies to assemble a model for FLC origins and development. Furthermore, we underscore the need for conclusive identification of the source population of fetal Leydig cells.
Collapse
Affiliation(s)
- S L Griswold
- Program in Developmental Biology, Baylor College of Medicine, Houston, Tex., USA
| | | |
Collapse
|
25
|
O'Shaughnessy PJ, Baker PJ, Monteiro A, Cassie S, Bhattacharya S, Fowler PA. Developmental changes in human fetal testicular cell numbers and messenger ribonucleic acid levels during the second trimester. J Clin Endocrinol Metab 2007; 92:4792-801. [PMID: 17848411 DOI: 10.1210/jc.2007-1690] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
CONTEXT Normal fetal testis development is essential for masculinization and subsequent adult fertility. The second trimester is a critical period of human testicular development and masculinization, but there is a paucity of reliable developmental data. OBJECTIVE The objective of the study was to analyze second-trimester human testicular morphology and function. DESIGN This was an observational study of second-trimester testis development. SETTING The study was conducted at the Universities of Glasgow and Aberdeen. PATIENTS/PARTICIPANTS Testes were collected from 57 morphologically normal fetuses of women undergoing elective termination of normally progressing pregnancies (11-19 wk gestation). MAIN OUTCOME MEASURE(S) Testicular morphology, cell numbers, and quantitative expression of 22 key testicular genes were determined. RESULTS Sertoli cell and germ cell number increased exponentially throughout the second trimester. Leydig cell number initially increased exponentially but slowed toward 19 wk. Transcripts encoding Sertoli (KITL, FGF9, SOX9, FSHR, WT1) and germ (CKIT, TFAP2C) cell-specific products increased per testis through the second trimester, but expression per cell was static apart from TFAP2C, which declined. Leydig cell transcripts (HSD17B3, CYP11A1, PTC1, CYP17, LHR, INSL3) also remained static per cell. Testicular expression of adrenal transcripts MC2R, CYP11B1, and CYP21 was detectable but unchanged. Expression of other transcripts known or postulated to be involved in testicular development (GATA4, GATA6, CXORF6, WNT2B, WNT4, WNT5A) increased significantly per testis during the second trimester. CONCLUSIONS The second trimester is essential for the establishment of Sertoli and germ cell numbers. Sertoli and Leydig cells are active throughout the period, but there is no evidence of changing transcript levels.
Collapse
Affiliation(s)
- P J O'Shaughnessy
- Division of Cell Sciences, University of Glasgow Veterinary School, Bearsden Road, Glasgow G61 1QH, United Kingdom. p.j.o'
| | | | | | | | | | | |
Collapse
|