51
|
Boucher E, Provost PR, Plante J, Tremblay Y. Androgen receptor and 17beta-HSD type 2 regulation in neonatal mouse lung development. Mol Cell Endocrinol 2009; 311:109-19. [PMID: 19576262 DOI: 10.1016/j.mce.2009.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 06/08/2009] [Accepted: 06/19/2009] [Indexed: 01/29/2023]
Abstract
A QPCR analysis of androgen receptor and several androgen metabolizing genes was performed during the saccular and alveolar stages of mouse lung development. Androgen receptor expression showed a statistically significant increase during the alveolar stage while levels of 17beta-hydroxysteroid dehydrogenase type 2 (17beta-HSD 2) expression significantly decreased at the end of the saccular stage and remained low throughout the alveolar period. 17beta-HSD 1, 17beta-HSD 5, 5alpha-reductase type 1, and mouse 3alpha-HSD did not present such a regulation. The androgen receptor protein was primarily detected in the nucleus of airway epithelial cells and of a subset of respiratory epithelial cells. 17beta-HSD 2 mRNA co-localized with androgen receptor protein during saccularization, but was absent from airway epithelium during alveolarization. Taken together, our results demonstrate temporal and spatial regulation of androgen receptor and 17beta-HSD 2 during the sacculo-alveolar transition period of mouse lung development suggesting control of androgen action.
Collapse
Affiliation(s)
- Eric Boucher
- Laboratory of Ontogeny and Reproduction, Centre Hospitalier Universitaire de Québec (CHUQ), Pavillon Centre Hospitalier de l'Université Laval (CHUL), Québec City, Québec, Canada
| | | | | | | |
Collapse
|
52
|
Chen H, Ge RS, Zirkin BR. Leydig cells: From stem cells to aging. Mol Cell Endocrinol 2009; 306:9-16. [PMID: 19481681 PMCID: PMC2749461 DOI: 10.1016/j.mce.2009.01.023] [Citation(s) in RCA: 197] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 01/22/2009] [Accepted: 01/23/2009] [Indexed: 12/27/2022]
Abstract
Leydig cells are the testosterone-producing cells of the testis. The adult Leydig cell population ultimately develops from undifferentiated mesenchymal-like stem cells present in the interstitial compartment of the neonatal testis. Four distinct stages of adult Leydig cell development have been identified and characterized: stem Leydig cells, progenitor Leydig cells, immature Leydig cells and adult Leydig cells. The stem Leydig cells are undifferentiated cells that are capable of indefinite self-renewal, differentiation, and replenishment of the Leydig cell niche. Progenitor Leydig cells are derived from the stem Leydig cells. These spindle-shaped cells are luteinizing hormone (LH) receptor positive, have high mitotic activity, and produce little testosterone but rather testosterone metabolites. The progenitor Leydig cells give rise to immature Leydig cells which are round, contain large amounts of smooth endoplasmic reticulum, and produce some testosterone but also very high levels of testosterone metabolites. A single division of these cells produces adult Leydig cells, which are terminally differentiated cells that produce high levels of testosterone. As men age, serum testosterone levels decline, and this is associated with alterations in body composition, energy level, muscle strength, physical, sexual and cognitive functions, and mood. In the Brown Norway rat, used extensively as a model for male reproductive aging, age-related reductions in serum testosterone result from significant decline in the ability of aged Leydig cells to produce testosterone in response to LH stimulation. This review describes Leydig cell development and aging. Additionally, the molecular mechanisms by which testosterone synthesis declines with aging are discussed.
Collapse
Affiliation(s)
- Haolin Chen
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
53
|
Lehraiki A, Racine C, Krust A, Habert R, Levacher C. Phthalates impair germ cell number in the mouse fetal testis by an androgen- and estrogen-independent mechanism. Toxicol Sci 2009; 111:372-82. [PMID: 19592451 PMCID: PMC2742583 DOI: 10.1093/toxsci/kfp153] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Data from experiments conducted almost exclusively in the rat have established that some phthalates have deleterious effects on the fetal testis probably due to their antiandrogenic and/or estrogenic effects, but their mechanisms of action remain unknown. A recent study reported that phthalates also have deleterious effects on human fetal testis with germ cell number, but not steroidogenesis altered. Therefore, we used organ culture of fetal testes at different stages of development to analyze the direct effects of phthalates on both steroidogenesis and gonocyte development and to determine if the effects of MEHP on these functions reported in the rat can be extended to other mammalian species. We defined specific periods of sensitivity of the fetal mouse testis to MEHP for these two functions and showed that the effects of phthalates on steroidogenesis vary with the developmental stage. Conversely, the strong deleterious effects of phthalates on germ cells were constantly present during the active phases of gonocyte development and thus share no relationship with the steroidogenic status. Moreover, all the effects of phthalates were unchanged in testes from mice deficient for estrogen (ERαKO or ERβKO) or androgen (Tfm) receptors. In conclusion, our results demonstrate that phthalates impair mouse fetal germ cell number similarly to other mammalian species, but are neither estrogenic nor antiandrogenic molecules because their effects do not involve, directly or indirectly, ER or AR.
Collapse
Affiliation(s)
- Abdelali Lehraiki
- Laboratory of Gonad Differentiation and Radiobiology, Stem Cells and Radiation Service, Institute of Cellular and Molecular Radiation Biology, Life Sciences Division, Commissariat à l'Energie Atomique, BP 6, 92265 Fontenay-aux-Roses, France
| | | | | | | | | |
Collapse
|
54
|
Welsh M, Sharpe RM, Walker M, Smith LB, Saunders PTK. New insights into the role of androgens in wolffian duct stabilization in male and female rodents. Endocrinology 2009; 150:2472-80. [PMID: 19131576 DOI: 10.1210/en.2008-0529] [Citation(s) in RCA: 37] [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: 11/19/2022]
Abstract
Androgen-mediated wolffian duct (WD) development is programmed between embryonic d 15.5 (e15.5) and 17.5 in male rats, and WD differentiation has been shown to be more susceptible to reduced androgen action than is its initial stabilization. We investigated regulation of these events by comparing fetal WD development at e15.5-postnatal d0 in male and female androgen receptor knockout mice, and in rats treated from e14.5 with flutamide (100 mg/kg/d) plus di-n(butyl) phthalate (500 mg/kg/d) to block both androgen action and production, testosterone propionate (20 mg/kg/d) to masculinize females, or vehicle control. In normal females, WD regression occurred by e15.5 in mice and e18.5 in rats, associated with a lack of epithelial cell proliferation and increased apoptosis, disintegration of the basement membrane, and reduced epithelial cell height. Exposure to testosterone masculinized female rats including stabilization and partial differentiation of WDs. Genetic or chemical ablation of androgen action in males prevented masculinization and induced WD regression via similar processes to those in normal females, except this occurred 2-3 d later than in females. These findings provide the first evidence that androgens may not be the only factor involved in determining WD fate. Other factors may promote survival of the WD in males or actively promote WD regression in females, suggesting sexually dimorphic differences in the preprogrammed setup of the WD.
Collapse
Affiliation(s)
- Michelle Welsh
- Human Reproductive Sciences Unit, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | | | | | | | | |
Collapse
|
55
|
Clewell RA, Kremer JJ, Williams CC, Campbell JL, Sochaski MA, Andersen ME, Borghoff SJ. Kinetics of selected di-n-butyl phthalate metabolites and fetal testosterone following repeated and single administration in pregnant rats. Toxicology 2009; 255:80-90. [DOI: 10.1016/j.tox.2008.10.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 09/26/2008] [Accepted: 10/15/2008] [Indexed: 11/28/2022]
|
56
|
Gore AC. Developmental programming and endocrine disruptor effects on reproductive neuroendocrine systems. Front Neuroendocrinol 2008; 29:358-74. [PMID: 18394690 PMCID: PMC2702520 DOI: 10.1016/j.yfrne.2008.02.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2007] [Revised: 02/21/2008] [Accepted: 02/22/2008] [Indexed: 01/01/2023]
Abstract
The ability of a species to reproduce successfully requires the careful orchestration of developmental processes during critical time points, particularly the late embryonic and early postnatal periods. This article begins with a brief presentation of the evidence for how gonadal steroid hormones exert these imprinting effects upon the morphology of sexually differentiated hypothalamic brain regions, the mechanisms underlying these effects, and their implications in adulthood. Then, I review the evidence that aberrant exposure to hormonally-active substances such as exogenous endocrine-disrupting chemicals (EDCs), may result in improper hypothalamic programming, thereby decreasing reproductive success in adulthood. The field of endocrine disruption has shed new light on the discipline of basic reproductive neuroendocrinology through studies on how early life exposures to EDCs may alter gene expression via non-genomic, epigenetic mechanisms, including DNA methylation and histone acetylation. Importantly, these effects may be transmitted to future generations if the germline is affected via transgenerational, epigenetic actions. By understanding the mechanisms by which natural hormones and xenobiotics affect reproductive neuroendocrine systems, we will gain a better understanding of normal developmental processes, as well as develop the potential ability to intervene when development is disrupted.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology and Toxicology, Institute for Neuroscience and Institute for Cellular and Molecular Biology, The University of Texas at Austin, 1 University Station A1915, Austin, TX 78712, USA.
| |
Collapse
|
57
|
Welsh M, Saunders PTK, Fisken M, Scott HM, Hutchison GR, Smith LB, Sharpe RM. Identification in rats of a programming window for reproductive tract masculinization, disruption of which leads to hypospadias and cryptorchidism. J Clin Invest 2008; 118:1479-90. [PMID: 18340380 DOI: 10.1172/jci34241] [Citation(s) in RCA: 544] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 01/23/2008] [Indexed: 01/18/2023] Open
Abstract
Becoming a phenotypic male is ultimately determined by androgen-induced masculinization. Disorders of fetal masculinization, resulting in hypospadias or cryptorchidism, are common, but their cause remains unclear. Together with the adult-onset disorders low sperm count and testicular cancer, they can constitute a testicular dysgenesis syndrome (TDS). Although masculinization is well studied, no unifying concept explains normal male reproductive development and its abnormalities, including TDS. We exposed rat fetuses to either anti-androgens or androgens and showed that masculinization of all reproductive tract tissues was programmed by androgen action during a common fetal programming window. This preceded morphological differentiation, when androgen action was, surprisingly, unnecessary. Only within the programming window did blocking androgen action induce hypospadias and cryptorchidism and altered penile length in male rats, all of which correlated with anogenital distance (AGD). Androgen-driven masculinization of females was also confined to the same programming window. This work has identified in rats a common programming window in which androgen action is essential for normal reproductive tract masculinization and has highlighted that measuring AGD in neonatal humans could provide a noninvasive method to predict neonatal and adult reproductive disorders. Based on the timings in rats, we believe the programming window in humans is likely to be 8-14 weeks of gestation.
Collapse
Affiliation(s)
- Michelle Welsh
- MRC Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, Edinburgh, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
58
|
Schlumpf M, Durrer S, Faass O, Ehnes C, Fuetsch M, Gaille C, Henseler M, Hofkamp L, Maerkel K, Reolon S, Timms B, Tresguerres JAF, Lichtensteiger W. Developmental toxicity of UV filters and environmental exposure: a review. ACTA ACUST UNITED AC 2008; 31:144-51. [DOI: 10.1111/j.1365-2605.2007.00856.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
59
|
Wu X, Wan S, Lee MM. Key factors in the regulation of fetal and postnatal Leydig cell development. J Cell Physiol 2008; 213:429-33. [PMID: 17674364 DOI: 10.1002/jcp.21231] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The primary function of testicular Leydig cells is the production of androgens to promote sexual differentiation in the fetus, secondary sexual maturation at puberty, and spermatogenesis in the adult. The fetal and postnatal (adult) populations of Leydig cells differ morphologically and have distinct profiles of gene expression. As postnatal Leydig cells differentiate, they transition through three discrete maturational stages characterized by decreasing proliferative rate and increasing testosterone biosynthetic capacity. In this review, we discuss the development of both fetal and postnatal Leydig cells and review the regulation of this process by some of the key hormones and growth factors.
Collapse
Affiliation(s)
- Xiufeng Wu
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | |
Collapse
|
60
|
Welsh M, Saunders PTK, Sharpe RM. The critical time window for androgen-dependent development of the Wolffian duct in the rat. Endocrinology 2007; 148:3185-95. [PMID: 17431008 DOI: 10.1210/en.2007-0028] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Androgens are thought to separately regulate stabilization and differentiation of the Wolffian duct (WD), but the time windows for these effects are unclear. To address this, fetal rats were exposed to flutamide within either an early window (EW) [embryonic day 15.5 (E15.5) to E17.5], when the WD degenerates in the female, or a later window (LW) (E19.5-E21.5), when the WD morphologically differentiates in the male, or during the full window of WD development (FW) (E15.5-21.5). WDs were examined for abnormalities during fetal (E21.5) or postnatal life, and anogenital distance and prostate presence/absence were recorded. Exposure to FW- or EW-flutamide, but not to LW-flutamide, induced comparable abnormalities in the fetal WD at E21.5, namely reduced WD coiling, reduced cell proliferation, reduced epithelial cell height, altered epithelial vimentin expression, and reduced expression of smooth muscle actin in the WD inner stroma. Exposure to EW- or FW-flutamide, but not to LW-flutamide, resulted in incomplete/absent WDs in more than 50% of males by adulthood, although such abnormalities were infrequent in fetal life. These findings suggest that androgen action during the EW is sufficient to promote WD morphological differentiation several days later. Because the androgen receptor is expressed in the WD stroma but not in the epithelium during this EW, WD differentiation is likely to be dependent on androgen-mediated signaling from the stroma to the epithelium. In conclusion, the critical window for androgen action in regulating WD development in the rat is between E15.5 and E17.5. This window is also important for prostate formation and anogenital distance masculinization.
Collapse
Affiliation(s)
- Michelle Welsh
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, United Kingdom.
| | | | | |
Collapse
|
61
|
Heckmann M, Hartmann MF, Kampschulte B, Gack H, Bödeker RH, Gortner L, Wudy SA. Persistent high activity of the fetal adrenal cortex in preterm infants: is there a clinical significance? J Pediatr Endocrinol Metab 2006; 19:1303-12. [PMID: 17220058 DOI: 10.1515/jpem.2006.19.11.1303] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In preterm infants, the activity of the fetal adrenal cortex continues until term. Dehydroepiandrosterone sulphate can block the synthesis of surfactant in vitro. The incidence of pulmonary disease is higher in male than in female preterm infants. OBJECTIVE To investigate the relationship between urinary excretion of fetal zone steroids (3beta-OH-5-ene-steroids) and the severity of lung disease in preterm infants with respect to gender. PATIENTS AND METHODS 3beta-OH-5-ene-steroids were profiled by gas chromatography-mass spectrometry in 24-h urinary samples in 61 preterm infants of less than 30 weeks gestational age. RESULTS The incidence of respiratory distress syndrome treated with surfactant in females (n = 30) was 47% and in males (n = 31) 71%, p = 0.07. Medians of total excretion rates of fetal zone steroids (microg/kg/d) in female (male) preterm infants were at day 1: 1,317 (895); day 2: 3,154 (7,723), p = 0.03; day 3: 5,502 (9,494), p = 0.08; day 5: 7,140 (10,407); week 2: 8,731 (9,720); week 3: 8,571 (10,079); week 4: 7,620 (7,825). Regression analysis did not reveal a significant influence of maximum excretion rates of fetal zone steroids or gender on the incidence of respiratory distress syndrome treated with surfactant. CONCLUSIONS Excretion rates of fetal zone steroids were 4-fold higher than previously reported indicating a persistent high activity of the fetal adrenal zone in preterm infants. Excretion rates of fetal zone steroids were significantly higher in male preterm infants compared to females at day 2 (trend at day 3) but did not have a significant influence on the incidence of respiratory distress syndrome.
Collapse
Affiliation(s)
- Matthias Heckmann
- Department of General Pediatrics and Neonatology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
62
|
Delbès G, Levacher C, Habert R. Estrogen effects on fetal and neonatal testicular development. Reproduction 2006; 132:527-38. [PMID: 17008464 DOI: 10.1530/rep.1.01231] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In recent years, evidences have accumulated that exposure to environmental components with estrogenic activity causes reproductive disorders in human populations. Studies conducted over the past 50 years have clearly shown a continual decline in semen quality accompanied by an increase in male reproductive disorders during this period in industrial countries. As healthy gametes are a prerequisite for healthy children, such disorders are a significant problem not only for the current society, but also for future generations. These male reproductive disorders have been attributed to xenobiotics, and particularly to xenoestrogens, which have steadily increased in diversity and concentration in the environment and food. Epidemiological, clinical, and experimental studies have suggested that excessive exposure to estrogens and xenoestrogens during fetal and neonatal development may induce testicular developmental disorders, leading to alterations in the adult male fertility. Recently, we have clearly demonstrated that fetal and neonatal testes are very sensitive to estrogens, as the inactivation of estrogen receptor α increases steroidogenesis and the inactivation of estrogen receptor β enhances development of the germ cell lineage in the male.
Collapse
|
63
|
Haavisto TE, Myllymäki SA, Adamsson NA, Brokken LJS, Viluksela M, Toppari J, Paranko J. The effects of maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on testicular steroidogenesis in infantile male rats. ACTA ACUST UNITED AC 2006; 29:313-22. [PMID: 16533353 DOI: 10.1111/j.1365-2605.2005.00568.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Exposure of adult male animals to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) decreases serum androgen concentrations. Reduction in androgen levels after maternal exposure has also been reported, but these results have not been reproduced. We have earlier shown that TCDD stimulates rather than inhibits testosterone synthesis in the prenatal rat testis. The aim of the present study was to elucidate in utero-induced effects of TCDD on testicular steroidogenesis in the 14-day-old infant rats. At that time the foetal Leydig cell population is still the prevailing source of androgens. Pregnant Sprague-Dawley dams were given a single oral dose of TCDD (0, 0.04, 0.2, or 1.0 microg/kg) on day 13 of pregnancy. On postnatal day 14, the body weight of male offspring was reduced after exposure to 1.0 microg/kg TCDD (from 33.9 +/- 1.66 g to 31.6 +/- 2.67 g). Relative testis weight, plasma testosterone, luteinizing hormone and follicle-stimulating hormone levels remained unaltered in all exposure groups. Moreover, in ex vivo incubations, testosterone and cAMP production was not affected. StAR protein level in the freshly isolated testes was increased in the 0.2 microg/kg group, and seminiferous cord diameter in the 0.04 microg/kg group. The present study confirms our earlier findings in in utero TCDD-exposed foetal testis indicating that maternal TCDD exposure does not negatively influence the developmental testosterone production of foetal type Leydig cells in rats.
Collapse
Affiliation(s)
- T E Haavisto
- Department of Biology, Laboratory of Animal Physiology, University of Turku, Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
64
|
Collins TFX, Sprando RL, Black TN, Olejnik N, Eppley RM, Alam HZ, Rorie J, Ruggles DI. Effects of zearalenone on in utero development in rats. Food Chem Toxicol 2006; 44:1455-65. [PMID: 16797818 DOI: 10.1016/j.fct.2006.04.015] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 04/14/2006] [Accepted: 04/27/2006] [Indexed: 12/31/2022]
Abstract
Zearalenone (ZE), an estrogenic mycotoxin produced by Fusarium graminearum or F. roseum, is one of the most common contaminants of cereal grains world-wide. The objective of this study was to determine the effects of ZE on in utero development of rats. Pregnant female Charles River Sprague-Dawley rats were gavaged once daily with ZE (in corn oil) at doses of 0, 1, 2, 4, or 8 mg/kg body weight on gestation days (GD) 6-19. All females survived to cesarean section on GD 20. At cesarean section, reproductive and developmental parameters were measured and blood was taken for hormone analysis. Dose-related decreases were seen in maternal feed consumption and body weight gain in all treated groups. Delayed fetal development was linked to maternal toxicity. Fetal body weight was significantly decreased in both sexes in all treated groups. ZE retarded skeletal ossification at 4 and 8 mg/kg. Fetal anogenital index (anogenital distance normalized for body weight) was increased in all treated groups, indicating an androgenic effect of ZE during fetal development. Fetal viability was significantly decreased at 8 mg/kg; significant decreases were observed in number of viable fetuses, and number of litters totally resorbed. At 4 and 8 mg/kg, maternal liver-body weight ratios were significantly increased and organ-brain weight ratios for weights of liver, heart, spleen, kidneys, and ovaries were significantly decreased. Gonadotropins (LH, FSH, and prolactin) and sex steroids (progesterone and estradiol) were analyzed from the blood serum obtained at cesarean section. LH in the 0, 1, 2, and 4 mg/kg groups showed minimal variation, and slightly increased at 8 mg/kg. FSH was decreased in the 1, 2, and 4 mg/kg groups, but the level at 8 mg/kg was slightly higher than the control level. Prolactin level was not affected at 1 mg/kg, slightly increased at 2 and 4 mg/kg, and significantly increased at 8 mg/kg. Progesterone was decreased at 2, 4, and 8 mg/kg and the decreases were significant at 2 and 4 mg/kg. Estradiol level was not affected at 1mg/kg, but dose-related decreases were observed at 2, 4, and 8 mg/kg. Only the 8 mg/kg level of estradiol was significantly decreased. In summary, ZE was maternally toxic and fetotoxic but not teratogenic. The increased anogenital distance observed in male and female fetuses was considered a hormonal change rather than a teratologic response. The increased anogenital distance indicated an androgenic effect. Based on the dose-related maternal and fetal toxicity in all treated groups, the NOEL for reproductive and teratogenic effects was less than 1 mg/kg.
Collapse
Affiliation(s)
- Thomas F X Collins
- Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, MD 20708, USA.
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Livera G, Delbes G, Pairault C, Rouiller-Fabre V, Habert R. Organotypic culture, a powerful model for studying rat and mouse fetal testis development. Cell Tissue Res 2006; 324:507-21. [PMID: 16520975 DOI: 10.1007/s00441-006-0167-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 01/09/2006] [Indexed: 11/29/2022]
Abstract
The key role of the fetal testis in the masculinization of genital organs has been known for a long time. More recently, the observed increases in male reproductive disorders has been postulated to be the result of changes in fetal and neonatal testis development in response to increasing environmental pollution. However, few tools are available for studying fetal testis development and the effects of physiological or toxic substances. We have developed an organ culture system in which rat fetal testis is grown on a filter floating on a synthetic medium containing no serum, hormones or biological factors. In this study, we have compared the long-term morpho-functional development of the various testicular cell types in this system with that observed in vivo and have extended this system to the mouse. Rat Leydig, Sertoli and germ cells and macrophages develop normally over a period of 1-2 weeks in this system. Fewer cells are produced than in vivo but the level of differentiated function is similar. Germ cells, which are difficult to culture in vitro, resume mitosis after a quiescent period, at the same time as in vivo. Similar results have been obtained with mouse fetuses, except that Leydig cells dedifferentiate in vitro if the testis is explanted after 13.5 days post conception. Testicular architecture and intercellular communication are sufficiently preserved for the development of the main fetal and neonatal testicular cell types in vitro with no added factors. Our floating-filter organotypic culture system in synthetic medium therefore allows the morpho-functional development of somatic and germ cells in fetal testis explants taken at all developmental stages in rat and at early stages in mouse. This method is potentially useful for studies of the effects of various factors, and of xenobiotics, in particular.
Collapse
Affiliation(s)
- Gabriel Livera
- Laboratory of Differentiation and Radiobiology of the Gonads, Research Unit on Gametogenesis and Genotoxicity, Université Paris 7, France.
| | | | | | | | | |
Collapse
|
66
|
Myllymäki SA, Haavisto TE, Brokken LJS, Viluksela M, Toppari J, Paranko J. In utero and lactational exposure to TCDD; steroidogenic outcomes differ in male and female rat pups. Toxicol Sci 2005; 88:534-44. [PMID: 16141434 DOI: 10.1093/toxsci/kfi308] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) has a potency to induce decreased fertility and structural reproductive anomalies in male and female mammals. While the activity profile of sex steroid hormone production distinctly differs in developing males and females, we wanted to analyze sex-specific effects of TCDD introduced in utero and via lactation on gonadal steroidogenesis and gonadotropin levels in male and female rat infant pups. One oral dose of TCDD (0, 0.04, 0.2, or 1.0 microg/kg) was given to dams on gestational day (GD) 13. Plasma testosterone, estradiol, progesterone, follicle stimulating hormone (FSH), luteinizing hormone (LH), and gonadal mRNA levels for steroid acute regulatory protein (StAR), cytochrome P-450 cholesterol side-chain cleavage (P450scc), 3beta-hydroxy-steroid-dehydrogenase/Delta(5)-Delta(4) isomerase type I (3beta-HSD1), P-450 17alpha-hydroxylase/17,20-lyase (P450-17alpha), and cytochrome P-450 aromatase (P450arom) were determined on postnatal days (PND) 10-16. TCDD 1.0 mug/kg reduced body weights but did not affect relative testis weight or alter testicular and ovarian histology. Plasma estradiol levels in dams and female pups were reduced on PND 14 and 16. Progesterone levels remained unaltered, and FSH levels were increased in female pups. In males, testosterone levels were elevated on PND 10. Gonadal mRNA levels for StAR and steroidogenic enzymes increased during the postnatal growth. TCDD caused no changes in relatively low testicular mRNA levels. However, significant reductions in StAR and P450arom mRNA levels were seen in PND 14 ovaries, and P450arom activity was decreased in isolated ovarian follicles. We conclude that developing testis and male gonadotropin secretion are resistant to TCDD-induced toxicity. In female pups, reduced estradiol, ovarian P450arom expression and enzyme activity levels, and elevated FSH levels may have a role in the development of ovarian dysfunction reported in TCDD-exposed females.
Collapse
Affiliation(s)
- S A Myllymäki
- Department of Biology, Laboratory of Animal Physiology, University of Turku, Finland
| | | | | | | | | | | |
Collapse
|
67
|
Delbès G, Levacher C, Duquenne C, Racine C, Pakarinen P, Habert R. Endogenous estrogens inhibit mouse fetal Leydig cell development via estrogen receptor alpha. Endocrinology 2005; 146:2454-61. [PMID: 15661855 DOI: 10.1210/en.2004-1540] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is now accepted that estrogens play a role in male fertility and that exposure to exogenous estrogens during fetal/neonatal life can lead to reproductive disorders in the male. However, the estrogen receptor (ER)-mediated processes involved in the regulation of male reproduction during fetal and neonatal development are still largely unclear. We previously reported that ER beta deficiency affects gametogenesis in mice but changes neither the number nor the differentiated functions of fetal Leydig cells. We show here that ER alpha-deficient mice (ER alpha-/-) display higher levels of testicular testosterone secretion than wild-type mice from fetal d 13.5 onwards. This results from higher levels of steroidogenic activity per fetal Leydig cell, as indicated by the hypertrophy of these cells and the higher levels of mRNA for StAR, P450c17 and P450scc in the testis, for a similar number of Leydig cells. Because LH is not produced on fetal d 13.5 and because no change in plasma LH concentration was observed in 2-d-old ER alpha-deficient mice, LH is probably not involved in the effects of estrogens on testicular steroidogenesis in fetal and early neonatal Leydig cells. Furthermore, inactivation of ER beta did not change the effect of ER alpha inactivation on steroidogenesis. Lastly, in an organ culture system, 1 mum diethylstilbestrol decreased the testosterone secretion of wild-type fetal and neonatal testes but not of ER alpha-/- testes. Thus, this study shows that endogenous estrogens physiologically inhibit steroidogenesis via ER alpha by acting directly on the testis early in fetal and neonatal development.
Collapse
Affiliation(s)
- Géraldine Delbès
- Unité de Gamétogenèse et Génotoxicité, Institut National de la Santé et de la Recherche Médicale Unité 566, Commissariat à l'Energie Atomique, Université Paris 7, Denis Diderot, Fontenay-aux-Roses, France
| | | | | | | | | | | |
Collapse
|
68
|
Delbès G, Levacher C, Pairault C, Racine C, Duquenne C, Krust A, Habert R. Estrogen receptor beta-mediated inhibition of male germ cell line development in mice by endogenous estrogens during perinatal life. Endocrinology 2004; 145:3395-403. [PMID: 15044378 DOI: 10.1210/en.2003-1479] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Epidemiological, clinical, and experimental studies have suggested that excessive exposure to estrogens during fetal/neonatal life can lead to reproductive disorders and sperm abnormalities in adulthood. However, it is unknown whether endogenous concentrations of estrogens affect the establishment of the male fetal germ cell lineage. We addressed this question by studying the testicular development of mice in which the estrogen receptor (ER) beta or the ERalpha gene was inactivated. The homozygous inactivation of ERbeta (ERbeta-/-) increased the number of gonocytes by 50% in 2- and 6-d-old neonates. The numbers of Sertoli and Leydig cells and the level of testicular testosterone production were unaffected, suggesting that estrogens act directly on the gonocytes. The increase in the number of gonocytes did not occur during fetal life but instead occurred just after birth, when gonocytes resumed mitosis and apoptosis. It seems to result from a decrease in the apoptosis rate evaluated by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling method and cleaved caspase-3 immunohistochemical detection. Last, mice heterozygous for the ERbeta gene inactivation behaved similarly to their ERbeta-/- littermates in terms of the number of gonocytes, apoptosis, and mitosis, suggesting that these cells are highly sensitive to the binding of estrogens to ERbeta. ERalpha inactivation had no effect on the number of neonatal gonocytes and Sertoli cells. In conclusion, this study provides the first demonstration that endogenous estrogens can physiologically inhibit germ cell growth in the male. This finding may have important implications concerning the potential action of environmental estrogens.
Collapse
Affiliation(s)
- Géraldine Delbès
- Unité de Gamétogenèse et Génotoxicité, Institut National de la Sante et de la Recherche Medicale Unite 566, Commissariat a l'Energie Atomique, Universite Paris 7-Denis Diderot, 92265 Fontenay-aux-Roses, France
| | | | | | | | | | | | | |
Collapse
|
69
|
Livera G, Pairault C, Lambrot R, Lelievre-Pegorier M, Saez JM, Habert R, Rouiller-Fabre V. Retinoid-Sensitive Steps in Steroidogenesis in Fetal and Neonatal Rat Testes: In Vitro and In Vivo Studies. Biol Reprod 2004; 70:1814-21. [PMID: 14960491 DOI: 10.1095/biolreprod.103.021451] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Retinoic acid (RA) was recently shown to modify testosterone secretion of the fetal testis in vitro. We characterized this effect by culturing rat testes explanted at various ages, from Fetal Day 14.5 to Postnatal Day 3. In basal medium, RA inhibited, in a dose-dependent manner, both basal and acute LH-stimulated testosterone secretion by testes explanted on Fetal Days 14.5, 15.5, and 16.5. It had no effect on testes from older animals. The negative effect of RA did not result from a diminution in the number of Leydig cells but from a decrease in P450c17 mRNA levels and in LH-stimulated cAMP production. However, the RA-induced decrease in P450C17 mRNA levels was also observed with neonatal testes, suggesting that this enzymatic step is no longer rate limiting at this developmental stage. To study the physiological relevance of RA effects, we used fetuses and neonates issued from mothers fed a vitamin A-deficient (VAD) diet, resulting in a threefold decrease of plasma retinol concentration. On Fetal Day 18.5 and on Posnatal Day 3, testosterone secretion by the testis ex vivo was significantly increased in VAD animals. This shows that the endogenous retinol inhibits differentiation and/or function of fetal Leydig cells before Fetal Day 18.5 and is required for the normal regression of fetal Leydig cell function that occurs after Fetal Day 18.5. In conclusion, our results show that retinoids play a negative role on the steroidogenic activity during the differentiation of rat fetal Leydig cells.
Collapse
Affiliation(s)
- G Livera
- INSERM U566-CEA-UNIVERSITE PARIS 7, CEA/DSV/DRR BP6, 92265 Fontenay aux Roses, France
| | | | | | | | | | | | | |
Collapse
|
70
|
Vega Matuszczyk J. Enhanced feminine sexual behavior and infertility in female rats prenatally treated with an antiestrogen. Scand J Psychol 2003; 44:251-6. [PMID: 12914588 DOI: 10.1111/1467-9450.00342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An attempt to elucidate the possible role of prenatal estrogen on the development of feminine sexual behavior and reproductive function was made by treating females with the antiestrogen CI628 prenatally on days 13-19. Control females were prenatally treated with saline or remained untreated. The animals were delivered by caesarian section on day 22 of pregnancy and placed with foster mothers whose newborn pups had been previously removed. Intact peripubertal females in each treatment group were observed for several reproductive measures, including the capacity to become pregnant. Other females were ovariectomized in adulthood and treated with estradiol benzoate (EB) (1, 1.5, 2 or 4 micro g/rat) and 0.5 mg progesterone and tested for receptivity, proceptivity and sexual partner preference. Two weeks after the completion of these tests, the females were injected daily for 7 days with 0.25 mg testosterone and tested for sexual partner preference and mounting behavior. The results obtained showed accelerated vaginal opening, and infertility in the antiestrogen-treated intact females and enhanced receptivity and proceptivity in response to 1 micro g EB in the antiestrogen ovariectomized females. Sexual partner preference and mounting behavior did not differ between groups. These results suggest an involvement of prenatal estrogen on the development of female reproductive function, but not on behavioral differentiation.
Collapse
Affiliation(s)
- Josefa Vega Matuszczyk
- Department of Behavioral Sciences, University Trollhättan-Uddevalla, Vänersborg, Sweden.
| |
Collapse
|
71
|
Shors TJ, Miesegaes G. Testosterone in utero and at birth dictates how stressful experience will affect learning in adulthood. Proc Natl Acad Sci U S A 2002; 99:13955-60. [PMID: 12359876 PMCID: PMC129804 DOI: 10.1073/pnas.202199999] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2002] [Indexed: 11/18/2022] Open
Abstract
Exposure to an acute stressful event can enhance learning in male rats, whereas exposure to the same event dramatically impairs performance in females. Here we tested whether the presence of sex hormones during early development organizes these opposite effects of stress on learning in males vs. females. In the first experiment, males were castrated at birth whereas females were injected with testosterone. Rats were trained as adults on the hippocampal-dependent learning task of trace eyeblink conditioning. Performance in adult males that had been castrated at birth was still enhanced by exposure to an acute stressful experience. However, adult females injected with testosterone at birth responded in the opposite direction, i.e., exposure to the stressor that typically reduces performance instead enhanced their levels of conditioning. In the second experiment, exposure to testosterone was manipulated in utero by injecting pregnant females with a testosterone antagonist. After foster rearing, adult offspring were exposed to the stressor and trained on the hippocampal-dependent learning task of trace conditioning. Although performance in adult females was unaffected by antagonizing testosterone in utero, i.e., stress still reduced performance, the enhancement of conditioning after stress in adult males was prevented. Thus, the presence of sex hormones during gestation and development organizes whether and how acute stressful experience will affect the ability to acquire new information in adulthood. As with many sexual behaviors, these cognitive responses to stress appear to be masculinized by exposure to testosterone and feminized by its absence during very early development.
Collapse
Affiliation(s)
- Tracey J Shors
- Department of Psychology and Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854-8020, USA.
| | | |
Collapse
|
72
|
Quadros PS, Goldstein AYN, De Vries GJ, Wagner CK. Regulation of sex differences in progesterone receptor expression in the medial preoptic nucleus of postnatal rats. J Neuroendocrinol 2002; 14:761-7. [PMID: 12372000 DOI: 10.1046/j.1365-2826.2002.00827.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The medial preoptic nucleus (MPN) of the rat, an excellent model for understanding the mechanisms involved in sexual differentiation, is highly sensitive to gonadal hormones during both pre- and post-natal life. Progesterone receptor (PR) expression is sexually dimorphic in the prenatal MPN. Males have significantly higher levels of PR-immunoreactivity (PRir) than females from approximately embryonic day 19 through at least the day of birth, suggesting that PR may play a role in sexual differentiation. Because the MPN is still sensitive to steroid hormones postnatally, the present study investigated PR expression in the MPN of males and females after birth using immunocytochemistry. Results indicate that a sex difference in PR expression persists until at least postnatal day (P) 28. However, females begin to express PR around P10. Because oestradiol regulates PR expression in the adult brain, this study also examined the influence of gonadal hormones on PR expression in the neonatal male and female MPN. Castration on the day of birth significantly reduced levels of PRir in the MPN by 24 h following surgery. Ovariectomy on P4, before the onset of ovarian steroidogenesis, prevented the induction of PR expression in the female MPN, observed in controls by P13. In both sexes, the presence of PRir in the MPN is dependent on gonadal hormone exposure. These findings suggest that differences in steroid secretion by the neonatal male and female gonads are responsible for producing sex differences in the level of PR expression in the postnatal MPN.
Collapse
Affiliation(s)
- P S Quadros
- Center for Neuroendocrine Studies and Neuroscience and Behaviour Program, University of Massachusetts, Amherst, MA, USA
| | | | | | | |
Collapse
|
73
|
Thin TH, Kim E, Yeh S, Sampson ER, Chen YT, Collins LL, Basavappa R, Chang C. Mutations in the helix 3 region of the androgen receptor abrogate ARA70 promotion of 17beta-estradiol-induced androgen receptor transactivation. J Biol Chem 2002; 277:36499-508. [PMID: 12068007 DOI: 10.1074/jbc.m202824200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The influence of estrogen on the development of the male reproductive system may be interrupted in a subset of partial androgen insensitivity syndrome (PAIS) patients. PAIS describes a wide range of male undermasculinization resulting from mutations in the androgen receptor (AR) or steroid metabolism enzymes that perturb androgen-AR regulation of male sex organ development. In this study, we are interested in determining if PAIS-derived AR mutants that respond normally to androgen have altered responses to estrogen in the presence of ARA70, a coregulator previously shown to enhance 17beta-estradiol E2-induced AR transactivation. The wild-type AR (wtAR) and two PAIS AR mutants, AR(S703G) and AR(E709K), all bind to androgen and E2 and subsequently translocate to the nucleus. Whereas ARA70 functionally interacts with the wtAR and the PAIS AR mutants in response to androgen, E2 only promotes the functional interaction between ARA70 and the wtAR but not the PAIS AR mutants. ARA70 increases E2 competitive binding to the wtAR in the presence of low level androgen and also retards E2 dissociation from the wtAR. ARA70 is present in both the cytoplasm and the nucleus of various mouse testicular cells during early embryogenesis day 16, at postpartum day 0 during estradiol synthesis and in the Leydig cells at postpartum day 49. ARA70 may be unable to modulate the PAIS AR mutants-E2 binding, diminishing the effect of E2 via AR during male reproductive system development in patients with such mutations. Therefore, the presence of ARA70 in the testosterone and E2-producing Leydig cells may enhance the overall activity of AR during critical stages of male sex organ development.
Collapse
Affiliation(s)
- Tin Htwe Thin
- George Whipple Laboratory for Cancer Research, Department of Pathology, University of Rochester, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Affiliation(s)
- R Habert
- INSERM-INRA U 418, Université Paris 7, 2 Place Jussieu, 75251, Paris, France
| | | | | |
Collapse
|
75
|
Haavisto T, Nurmela K, Pohjanvirta R, Huuskonen H, El-Gehani F, Paranko J. Prenatal testosterone and luteinizing hormone levels in male rats exposed during pregnancy to 2,3,7,8-tetrachlorodibenzo-p-dioxin and diethylstilbestrol. Mol Cell Endocrinol 2001; 178:169-79. [PMID: 11403907 DOI: 10.1016/s0303-7207(01)00425-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Changes in the perinatal testosterone surge have been related to demasculinization of the central nervous system and androgen-dependent growth of the reproductive organs in male mammals. Earlier reports suggest that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) interferes with androgen production, but the perinatal effects have remained elusive. In the present study we explored in utero-effects of TCDD (0.05, 0.1, 0.5, and 1.0 microg/kg), introduced on day 13.5 of pregnancy, on prenatal (day 19.5 post-conception [p.c.]) testosterone (T) surge and pituitary luteinizing hormone (LH) production in TCDD-resistant Han/Wistar (H/W) and TCDD-sensitive Long-Evans (L-E) rats. To elucidate estrogenic effects on T and LH production, Sprague-Dawley (S-D) fetuses with previously known DES-sensitivity were exposed in utero to diethylstilbestrol (DES, 100-300 microg/kg) on days 13.5, 15.5, and 17.5 p.c. For comparison, H/W fetuses that responded to TCDD treatments were exposed to DES at concentration of 100 microg/kg. It was found that TCDD has a stimulatory effect on testicular T synthesis in the H/W fetuses and that their circulating T concentrations increased significantly. The effect was not seen in the inbred L-E fetuses, which throughout the study showed considerably low testicular T levels. Pituitary LH concentrations also increased in the H/W fetuses exposed to TCDD. Effects of TCDD (1.0 microg/kg) in the H/W fetuses could be confirmed in vitro by human chorionic gonadotropin (hCG) stimulation assay showing the highest response rate in the TCDD exposed testes. Stimulation of cyclic AMP (adenosine-3', 5'-cyclic monophosphate[cAMP]) production was not considerably altered by in utero TCDD exposure. A significant depression in testicular and plasma T content was seen in the DES-exposed S-D and H/W fetuses, but pituitary LH levels did not alter considerably. In the presence of hCG, DES-exposed testes showed lower in vitro T and cAMP production rates compared to the untreated testes. TCDD (1.0 microg/kg) increased and DES decreased the male body weight gain, but the changes were not sex-dependent. It is concluded that TCDD may increase the amplitude of the prenatal testosterone surge in male rats by stimulating pituitary LH production and enhancing the sensitivity of the fetal testis to LH. DES, on the contrary, apparently impairs testicular steroidogenesis and pituitary function.
Collapse
Affiliation(s)
- T Haavisto
- Laboratory of Animal Physiology, Department of Biology, University of Turku, 20014, Turku, Finland
| | | | | | | | | | | |
Collapse
|
76
|
Livera G, Rouiller-Fabre V, Habert R. Retinoid receptors involved in the effects of retinoic acid on rat testis development. Biol Reprod 2001; 64:1307-14. [PMID: 11319133 DOI: 10.1095/biolreprod64.5.1307] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We have previously shown that retinoic acid (RA) is able to act on the development of Leydig, Sertoli, and germ cells in the testis in culture (Livera et al., Biol Reprod 2000; 62:1303-1314). To identify which receptors mediate these effects, we have now added selective agonists and antagonists of retinoic acid receptors (RARs) or retinoid X receptors (RXRs) in the same organotypic culture system. The RAR alpha agonist mimicked most of the effects of RA on the cultured fetal or neonatal testis, whereas the RAR beta, gamma, and pan RXR agonists did not. The RAR alpha agonist decreased the testosterone production, the number of gonocytes, and the cAMP response to FSH of fetal testis explanted at 14.5 days postconception (dpc). The RAR alpha agonist disorganized the cords of the 14.5-dpc cultured testis and increased the cord diameter in cultured 3-days-postpartum (dpp) testis in the same way as RA. All these RA effects could be reversed by an RAR alpha antagonist and were unchanged by an RAR beta/gamma antagonist. The RAR beta agonist, however, increased Sertoli cell proliferation in the 3-dpp testis in the same way as RA, and this effect was blocked by an RAR beta antagonist. The RAR gamma and the pan RXR agonists had no selective effect. These results suggest that all the effects of RA on development of the fetal and neonatal testis are mediated via RAR alpha, except for its effect on Sertoli cell proliferation, which involves RAR beta.
Collapse
Affiliation(s)
- G Livera
- Université Paris 7 and INSERM-INRA U 418, Tour 33/43, case 7126, 75251 Paris Cedex 05, France
| | | | | |
Collapse
|
77
|
Migrenne S, Pairault C, Racine C, Livera G, Géloso A, Habert R. Luteinizing hormone-dependent activity and luteinizing hormone-independent differentiation of rat fetal Leydig cells. Mol Cell Endocrinol 2001; 172:193-202. [PMID: 11165053 DOI: 10.1016/s0303-7207(00)00339-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Addition of 5x10(-2) U/ml recombinant luteinizing hormone (LH) to testes from fetuses at 16.5 day post conception (dpc) cultured for 5 days increased the number of Leydig cells by 34% and the acute LH-stimulated testosterone production by 600%. To determine whether these positive effects of LH in vitro are physiologically relevant in vivo, fetuses were decapitated on days 16.5 pc (before the onset of LH expression in the hypophysis) or 18.5 pc (before the surge of LH in the fetal plasma) and removed at 21.5 dpc. The number of fetal Leydig cells per testis and the acute LH-stimulated testosterone production by the testes ex vivo were unaltered by decapitation. Since, in all groups, the number of Leydig cells doubled between 16.5 and 18.5 dpc and between 18.5 and 21.5 dpc, these results suggest that neither the appearance of new fully differentiated fetal Leydig cells nor the maintenance of differentiated functions in existing fetal Leydig cells depend on LH during late fetal life, although this hormone is present in the plasma. Decapitation reduced the testosterone concentrations in the plasma (-56%) and in the testis in vivo (-67%) and the basal testosterone secretion of the testis ex vivo (-70%). This suggests that LH is required to maintain the physiological activity of the Leydig cell during late fetal life. However, the decrease of the in vivo testosterone production after decapitation was not sufficient to impair the growth of the Wolffian ducts and the lengthening of the anogenital distance. In conclusion, during late fetal life in the rat, Leydig cells are LH-independent for their functional differentiation and LH-dependent for their activity.
Collapse
Affiliation(s)
- S Migrenne
- INSERM-INRA U 418, Tour 331443, Case 7126, Université Paris 7, 2 Place Jussieu, 75251 Cedex 05, Paris, France
| | | | | | | | | | | |
Collapse
|
78
|
Nuñez JL, Jurgens HA, Juraska JM. Androgens reduce cell death in the developing rat visual cortex. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2000; 125:83-8. [PMID: 11154764 DOI: 10.1016/s0165-3806(00)00126-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We have previously shown that males have more neurons than females in the primary visual cortex, and neonatal androgens play an important role in this difference. Also, we have found that females experience more cell death during development in this region than in males. Therefore, we hypothesized that the neonatal hormone environment directly influences the amount of cell death. In the present experiment, female rats were implanted with dihydrotestosterone (DHT) or estradiol at postnatal day 1. These animals, along with control males and females, were sacrificed on postnatal days 6, 11 and 25. Using unbiased stereology to quantify neuronal and pyknotic cell density, we observed that females implanted with DHT had a similar pattern and proportion of cells dying as control males. Additionally, developmental cell death in females implanted with estradiol was not significantly different than control females. Thus, neonatal androgens have an inhibitory effect on developmental cell death in the rat primary visual cortex.
Collapse
Affiliation(s)
- J L Nuñez
- Neuroscience Program and Department of Psychology, University of Illinois at Champaign-Urbana, Champaign, IL 61820, USA
| | | | | |
Collapse
|
79
|
Abstract
Immunocytochemical analysis of endocrine cells of the developing rat pituitary has shown clearly that, starting from E16, the galanin-immunoreactive cells are heterogeneous, the neuropeptide being present in cells containing the majority of pituitary hormones. A proportion of galanin-containing corticotropes, starting from about 17% at E16 reached about 29% at E21, while the galanin-containing lactotrope increase in the same period was from about 13% to 34%. Galanin-containing thyrotropes ranged from about 11% at E16 to about 13% at E21, while the proportion of galanin-containing luteotropes increased consistently from about 17% at E19 to about 27% at E21. Only the trend of galanin-containing somatotropes was inverted, falling from about 26% to 13%. We conclude that the multiple coexistence of galanin with most of the pituitary hormones during fetal development is a further example of plasticity of endocrine pituitary cells, and that galanin may have a role in cytodifferentiation.
Collapse
Affiliation(s)
- V Cimini
- Dipartimento di Scienze Biomorfologiche e Funzionali, Cattedra di Istologia, Via Pansini 5, 80131, Napoli, Italy.
| |
Collapse
|
80
|
Livera G, Rouiller-Fabre V, Durand P, Habert R. Multiple effects of retinoids on the development of Sertoli, germ, and Leydig cells of fetal and neonatal rat testis in culture. Biol Reprod 2000; 62:1303-14. [PMID: 10775181 DOI: 10.1095/biolreprod62.5.1303] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
We investigated the effect of retinoids on the development of Sertoli, germ, and Leydig cells using 3-day culture of testes from fetuses 14.5 and 18.5 days post-conception (dpc) and from neonates 3 days postpartum (dpp). Addition of 10(-6) M and 3.10(-8) M retinoic acid (RA) caused a dose-dependent disruption of the seminiferous cords in 14.5-day-old fetal testes, without any change in the 5-bromo-2'-deoxyuridine (BrdU) labeling index of the Sertoli cells. RA caused no disorganization of older testes, but it did cause hyperplasia of the Sertoli cells in 3-dpp testes. Fragmentation of the Sertoli cell DNA was not detected in control or RA-treated testes at any age studied. The cAMP produced in response to FSH was significantly decreased in RA-treated testes for all studied ages. Both 10(-6) M and 3.10(-8) M RA dramatically reduced the number of gonocytes per 14.5-dpc testis. This resulted from a high increase in apoptosis, which greatly exceeded the slight increase of mitosis. RA caused no change in the number of gonocytes in testes explanted on 18.5 dpc (the quiescent period), whereas it increased this number in testes explanted on 3 dpp (i.e., when gonocyte mitosis and apoptosis resume). Lastly, RA and retinol (RE) reduced both basal and acute LH-stimulated testosterone secretion by 14.5-dpc testis explants, without change in the number of 3beta-hydroxysteroid dehydrogenase-positive cells per testis. Retinoids had no effect on basal or LH-stimulated testosterone production by older testes. In conclusion, RE and RA are potential regulators of the development of the testis and act mainly negatively during fetal life and positively during the neonatal period on the parameters we have studied.
Collapse
Affiliation(s)
- G Livera
- Université Paris 7 and INSERM-INRA U 418, Tour 33/43, case 7126, 75251 Paris Cedex 05, France
| | | | | | | |
Collapse
|
81
|
Affiliation(s)
- M P Hedger
- Monash University Institute of Reproduction and Development, Monash Medical Centre, Clayton, Victoria, Australia
| | | |
Collapse
|
82
|
Boulogne B, Olaso R, Levacher C, Durand P, Habert R. Apoptosis and mitosis in gonocytes of the rat testis during foetal and neonatal development. INTERNATIONAL JOURNAL OF ANDROLOGY 1999; 22:356-65. [PMID: 10624604 DOI: 10.1046/j.1365-2605.1999.00191.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study was undertaken to determine the extent of apoptosis and mitosis in the various testicular cell types throughout rat development from foetal day 14.5 to postnatal days 9-10. Apoptotic activity was studied by detecting DNA fragmentation (TUNEL method) in situ. A TUNEL-positive reaction was detected in gonocytes, while none of the other testicular cells were labelled. The morphology of the TUNEL-positive gonocytes was characteristic of apoptotic cells and was different from that observed in experimentally induced necrosis. The percentage of stained gonocytes peaked on day 15.5-16.5 post-conception (dpc), decreased thereafter and no TUNEL-positive gonocytes were found from foetal day 18.5 onwards. On postnatal day 2, apoptosis resumed and increased to reach a maximum on day 7. Mitosis in the gonocytes, as evaluated by the immunodetection of 5-bromo-2'-deoxyuridine (BrdU) incorporation, was present during the same developmental periods but the ratio of BrdU-positive/TUNEL-positive gonocytes was much greater in the foetal period than in the neonatal period. In an organotypic culture system, the changes in the apoptotic and mitotic activities of the gonocytes in testicular explants from foetuses on days 18.5 and 20.5 or from neonates on day 3, cultured for two days were similar to those observed in vivo. Addition of LH or FSH did not influence either apoptosis or mitosis in the germ cells. These results suggest that both apoptosis and mitosis of gonocytes are independent of gonadotrophins and are mainly controlled by intratesticular factors.
Collapse
Affiliation(s)
- B Boulogne
- INSERM-INRA U 418, Université Paris 7, France
| | | | | | | | | |
Collapse
|
83
|
Seuntjens E, Vankelecom H, Quaegebeur A, Vande Vijver V, Denef C. Targeted ablation of gonadotrophs in transgenic mice affects embryonic development of lactotrophs. Mol Cell Endocrinol 1999; 150:129-39. [PMID: 10411307 DOI: 10.1016/s0303-7207(99)00011-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Ablation of pituitary gonadotrophs was obtained in transgenic mice expressing diphtheria toxin A (DTA) under control of the -313/+48 bovine glycoprotein hormone alpha-subunit (alphaSU) promoter, previously shown to be active in mouse gonadotrophs but not in thyrotrophs. Development of hormone-producing cell types was assessed on the day of birth by computer-assisted image analysis on paraffin-embedded, immunostained pituitary sections. Six out of 50 transgenic F0 ('founder') mice (3 males and 3 females) showed a nearly complete disappearance of gonadotrophs but not of thyrotrophs. The number of lactotrophs and the relative area occupied by PRL-immunoreactivity were significantly reduced in the gonadotroph-depleted mice. The size of lactotroph clusters was smaller in the absence of gonadotrophs. The number and immunoreactive area of corticotrophs and somatotrophs, on the other hand, were not significantly affected by gonadotroph ablation. Based on the reported evidence that fetal ovaries do not produce steroid hormones as a result of lack of expression of at least three of the steroidogenic enzymes, P450scc, P450c17, and P450arom, the present observations can hardly be explained by a decline in estrogen levels due to gonadotroph ablation. Rather, the present data indicate that gonadotrophs directly stimulate the development of lactotrophs during fetal and early postnatal life, consistent with previous in vitro observations, and/or that gonadotrophs may share a cell-lineage relationship with a subpopulation of lactotrophs.
Collapse
Affiliation(s)
- E Seuntjens
- Department of Molecular Cell Biology, University of Leuven, Medical School, Belgium
| | | | | | | | | |
Collapse
|
84
|
Cook JC, Klinefelter GR, Hardisty JF, Sharpe RM, Foster PM. Rodent Leydig cell tumorigenesis: a review of the physiology, pathology, mechanisms, and relevance to humans. Crit Rev Toxicol 1999; 29:169-261. [PMID: 10213111 DOI: 10.1080/10408449991349203] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Leydig cells (LCs) are the cells of the testis that have as their primary function the production of testosterone. LCs are a common target of compounds tested in rodent carcinogenicity bioassays. The number of reviews on Leydig cell tumors (LCTs) has increased in recent years because of its common occurrence in rodent bioassays and the importance in assessing the relevance of this tumor type to humans. To date, there have been no comprehensive reviews to identify all the compounds that have been shown to induce LCTs in rodents or has any review systematically evaluated the epidemiology data to determine whether humans were at increased risk for developing LCTs from exposure to these agents. This review attempts to fill these deficiencies in the literature by comparing the cytology and ontogeny of the LC, as well as the endocrine and paracrine regulation of both normal and tumorigenic LCs. In addition, the pathology of LCTs in rodents and humans is compared, compounds that induce LC hyperplasia or tumors are enumerated, and the human relevance of chemical-induced LCTs is discussed. There are plausible mechanisms for the chemical induction of LCTs, as typified by agonists of estrogen, gonadotropin releasing hormone (GnRH), and dopamine receptors, androgen receptor antagonists, and inhibitors of 5alpha-reductase, testosterone biosynthesis, and aromatase. Most of these ultimately involve elevation in serum luteinizing hormone (LH) and/or LC responsiveness to LH as proximate mediators. It is expected that further work will uncover additional mechanisms by which LCTs may arise, especially the role of growth factors in modulating LC tumorigenesis. Regarding human relevance, the pathways for regulation of the hypothalamo-pituitary-testis (HPT) axis of rats and humans are similar, such that compounds that either decrease testosterone or estradiol levels or their recognition will increase LH levels. Hence, compounds that induce LCTs in rats by disruption of the HPT axis pose a risk to human health, except for possibly two classes of compounds (GnRH and dopamine agonists). Because GnRH and prolactin receptors are either not expressed or are expressed at very low levels in the testes in humans, the induction of LCTs in rats by GnRH and dopamine agonists would appear not to be relevant to humans; however, the potential relevance to humans of the remaining five pathways of LCT induction cannot be ruled out. Therefore, the central issue becomes what is the relative sensitivity between rat and human LCs in their response to increased LH levels; specifically, is the proliferative stimulus initiated by increased levels of LH attenuated, similar, or enhanced in human vs. rat LCs? There are several lines of evidence that suggest that human LCs are quantitatively less sensitive than rats in their proliferative response to LH, and hence in their sensitivity to chemically induced LCTs. This evidence includes the following: (1) the human incidence of LCTs is much lower than in rodents even when corrected for detection bias; (2) several comparative differences exist between rat and human LCs that may contribute, at least in part, to the greater susceptibility of the rat to both spontaneous and xenobiotic-induced LCTs; (3) endocrine disease states in man (such as androgen-insensitivity syndrome and familial male precocious puberty) underscore the marked comparative differences that exist between rats and man in the responsiveness of their LC's to proliferative stimuli; and (4) several human epidemiology studies are available on a number of compounds that induce LCTs in rats (1,3-butadiene, cadmium, ethanol, lactose, lead, nicotine) that demonstrate no association between human exposure to these compounds and induction of LC hyperplasia or adenomas. (ABSTRACT TRUNCATED)
Collapse
Affiliation(s)
- J C Cook
- DuPont Haskell Laboratory, Newark, DE, USA
| | | | | | | | | |
Collapse
|
85
|
Butler CM, Harry JL, Deakin JE, Cooper DW, Renfree MB. Developmental expression of the androgen receptor during virilization of the urogenital system of a marsupial. Biol Reprod 1998; 59:725-32. [PMID: 9746719 DOI: 10.1095/biolreprod59.4.725] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In the marsupial tammar wallaby, virilization begins approximately 3 wk after the onset of testosterone synthesis. In the eutherian mammal, in contrast, the onset of virilization of the male urogenital tract occurs shortly after the onset of androgen synthesis. Androgen action requires the presence of the androgen receptor to mediate a response in target tissues. We therefore investigated the developmental expression of the androgen receptor (AR) in both sexes of the tammar wallaby. AR gene transcript was detected in fetal gonad and brain as early as Day 19 of the 26.5-day gestation, 7 days earlier than the first rise in testicular testosterone (Days 0-5 postpartum [p.p.]). Immunoreactive AR was identified in the male urogenital sinus (UGS) 2 days before birth and in the female UGS and mammary glands by the day of birth. AR was present in the UGS, vagina, and prostate until Day 152 p.p., the oldest age examined. AR was identified in the gubernaculum testis at Day 2 p.p. and became more abundant by Day 32. In the phallus of both sexes, AR was identified by Day 4 p.p. and until Day 157, the oldest age examined. AR was not detected in the scrotum at any age from the day of birth to Day 157. Maturation of the phallus, wolffian duct, and epididymis was marked by appearance of epithelial immunostaining. AR was localized in the epithelium of the UGS in females by Day 50 p.p. but was not found in the epithelium of the male UGS up to Day 152 p.p., the oldest examined. AR were found in the mesenchyme of the UGS of male and female tammars 3-4 wk before virilization is first evident in the male at Day 25 p.p. We conclude that the presence of AR is not the initiating signal for virilization of the UGS in this marsupial male.
Collapse
Affiliation(s)
- C M Butler
- Department of Zoology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | | | | | | | | |
Collapse
|
86
|
Sweezey NB, Ghibu F, Gagnon S, Schotman E, Hamid Q. Glucocorticoid receptor mRNA and protein in fetal rat lung in vivo: modulation by glucocorticoid and androgen. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:L103-9. [PMID: 9688941 DOI: 10.1152/ajplung.1998.275.1.l103] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pulmonary glucocorticoid receptor (GR) is essential to timely preparation for the onset of breathing air at birth. We have previously used primary culture of late-gestation fetal rat lung cells to demonstrate differential regulation of GR by glucocorticoid depending on cell type. In this study, we hypothesized that the action of glucocorticoid on GR mRNA expression and protein elaboration in lung cells might be modulated by interactions present in vivo but not in primary culture. Given that male sex hormone (androgen) has an inhibitory effect on antenatal lung development, we also postulated that androgen would decrease antenatal lung GR. We report that antenatal maternal injection of the glucocorticoid dexamethasone (1 mg/kg) enhanced fetal lung cellular levels of GR mRNA and protein as assessed by in situ hybridization and immunocytochemistry (ICC), respectively. ICC was performed using polyclonal rabbit anti-human antibody that reacts with rat GR whether bound to ligand or not and does not interfere with GR binding to DNA. Levels of GR mRNA and protein were enhanced in cells throughout all areas of the lung tissue, suggesting that interactions occurring in intact tissue may override the previously reported direct inhibition by glucocorticoid of GR protein elaboration in isolated fetal rat lung epithelial cells. Furthermore, antenatal administration of the androgen 5alpha-dihydrotestosterone (0.2 mg/kg) reduced tissue levels of GR mRNA and protein, consistent with androgenic inhibition of antenatal lung development by decreasing GR. We conclude that glucocorticoids and androgens exert opposite effects on fetal lung GR.
Collapse
Affiliation(s)
- N B Sweezey
- Respiratory Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | |
Collapse
|
87
|
Rouiller-Fabre V, Lecref L, Gautier C, Saez JM, Habert R. Expression and effect of insulin-like growth factor I on rat fetal Leydig cell function and differentiation. Endocrinology 1998; 139:2926-34. [PMID: 9607803 DOI: 10.1210/endo.139.6.6035] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Insulin like growth factor I (IGF-I) is believed to be a potent para/autocrine stimulator of Leydig cell function in adult testis. We investigated whether IGF-I is also an intratesticular regulator of fetal Leydig cell function by measuring its production in the fetal testis and its ability to affect testicular steroidogenesis during fetal development. Northern blot analysis revealed one major IGF-I transcript of 7-7.5 kb and two minor transcripts of 3.8 and 1.8 kb in 20.5 day fetal testis. IGF-I was detected by RIA in 16.5 fetal day testes, and the amounts of IGF-I secreted by 16.5 and 20.5 fetal day testes in vitro were much greater than the amounts contained in the testes, indicating active synthesis in culture. The secretion of IGF-I by the fetal testis in vitro was increased with testicular age and time in culture. It was not modified by gonadotropins or (Bu)2cAMP. Testosterone secretion by fetal testes explanted 13.5, 16.5, 18.5, and 20.5 days after conception and cultured in the presence or absence of 100 ng/ml LH for 3 days was not affected by the addition of 50 ng/ml IGF-I to the medium. In contrast, the addition of IGF-I to dispersed fetal testicular cells cultured for 3 days in the presence or absence of LH increased the number of Leydig cells identified by a positive cytochemical reaction for 3beta-hydroxysteroid dehydrogenase (3betaHSD). This was more pronounced with cells from 16.5- day-old fetuses (stage when the fetal Leydig cells are differentiating in vivo) than with 20.5-day-old fetuses cells (stage when the number and the function of fetal Leydig cells are stable or decreasing). It results from both an increased differentiation of mesenchymal cells in fetal Leydig cells and an increase in the mitotic index of the fetal Leydig cells, as inferred from the small increase in the percentage of bromodeoxyuridine/3betaHSD-positive cells. Both LH and IGF-I increased significantly testosterone production by day 16.5 cells. In the presence of LH, a high amount of testosterone was produced per 3betaHSD-positive cell; IGF-I further increased this production. This effect was not observed with day 20.5 cells. The amounts of testosterone produced per 3betaHSD-positive cell cultured in the presence of both LH and IGF-I were more than additive. Like IGF-I, insulin (50 ng/ml) increased testosterone secretion per 3betaHSD-positive cells in cultures of day 16.5 cells, but not in those of day 20.5, cells. Lastly, IGF-I also increased the steroidogenic activity of each Leydig cell in cultures containing (Bu)2cAMP, but its effects were weaker than those observed in the presence of LH. This suggests that IGF-I has sites of action both upstream and downstream cAMP generation. These results suggest that IGF-I acts as paracrine/autocrine factor in the differentiation and activity of fetal Leydig cells.
Collapse
|
88
|
Gautier C, Levacher C, Saez JM, Habert R. Transforming growth factor beta1 inhibits steroidogenesis in dispersed fetal testicular cells in culture. Mol Cell Endocrinol 1997; 131:21-30. [PMID: 9256360 DOI: 10.1016/s0303-7207(97)00087-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
TGF beta1 has been detected by immunohistochemistry in the rat fetal testis. Therefore, we attempted to determine whether this factor can act as a local regulator of Leydig cell function during fetal development. An inhibitory effect of TGF beta1 on basal and luteinizing hormone (LH)-stimulated testosterone secretion by fetal testes in vitro was observed only with testes from 13.5 day-old fetuses and not with testes from older stages. The lack of effect of exogenous TGF beta1 in organ culture after day 13.5 might be related to an elevated intratesticular concentration that would already exert maximal biological effect. On the contrary, in a model of dispersed testicular cells in culture, TGF beta1 was able to inhibit LH-stimulated testosterone production by fetal Leydig cells from 16.5 and 20.5 day-old fetuses. This inhibition of LH-stimulated testosterone production was dose- and time-dependent and was maximal after 48 h of treatment with 1 ng/ml TGF beta1, with testosterone secretion being reduced to 25% of control values. Inhibition of testosterone secretion was also observed in basal and dbcAMP-stimulated conditions, suggesting that one site of action of TGF beta1 is located after the production of cAMP. However, TGF beta1 was also able to inhibit LH-induced cAMP production. As demonstrated by the transformation of steroidogenic precursors into testosterone, TGF beta1 did not significantly alter 3beta-hydroxysteroid dehydrogenase (3beta HSD) activity but induced a strong inhibition of cytochrome P450 17alpha-hydroxylase/C17-20 lyase (P450C17) activity which was associated with a marked diminution of cytochrome P450C17 mRNA levels (26% of control values) but not of cytochrome P450scc mRNA. In addition to its effect on steroidogenesis, TGF beta1 exhibited morphogenic actions on the fetal testicular cells, inducing spreading when the cells were adherent and aggregation when the cells were cultured in conditions of lesser adherence and without any significant effect on either total cell number or 3beta HSD positive cells. Taken together these results suggest that TGF beta1 likely plays a morphogenic and physiological role very early in the fetal testis via paracrine/autocrine mechanisms.
Collapse
Affiliation(s)
- C Gautier
- INSERM-INRA U 418 Université Paris, France
| | | | | | | |
Collapse
|
89
|
Vega Matuszczyk J, Appa RS, Larsson K. Postnatal testicular secretions partially restore the disturbances in reproductive activity caused by prenatal hormonal manipulation. Physiol Behav 1997; 62:137-43. [PMID: 9226353 DOI: 10.1016/s0031-9384(97)00020-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Male rats that prenatally had been exposed to an antiestrogen, nitromifene citrate (CI 628), showed evidence of impaired defeminization and masculinization in adulthood, suggesting a role of endogenous estrogen for the sexual differentiation of the male. The present study was undertaken to investigate a possible role of postnatal testicular secretions for the above behavioral effects. Male rats were exposed prenatally to CI 628 (1 mg/rat) or saline, and castrated on Day 0, Day 10, or Day 90 after birth. After treatment with gonadal hormones in adulthood, the males were tested for feminine and masculine sexual behavior and for sexual orientation, both when sexually naive and after they had acquired sexual experience. The following conclusions were drawn: 1. Permanent deficits of lordotic behavior were observed in all experimental groups, suggesting the importance of prenatal estrogen for the defeminization process. 2. Hop/darting and ear wiggling behaviors were enhanced in Day-0 and Day-10 castrates, and blocked in the Day-90 castrates. The restitution of these behaviors to normal levels in Day-90 castrates suggests that, in addition to prenatal estrogen, postnatal testicular secretions also are involved in the behavioral defeminization process. 3. Prenatal estrogen contributes to masculinization as evidenced by the impaired ejaculatory behavior observed in all experimental groups. 4. Male-typical sexual orientation toward the female seems to be facilitated by prenatal estrogen. Both the masculinization and the defeminization of male-typical sexual orientation toward a female were impaired by castration at birth and at Day 10 in the experimental animals, but a full restoration of the sexual orientation toward females was seen in Day-90 castrates, suggesting the restorative role of postnatal testicular secretions for these processes.
Collapse
|
90
|
Rajkowski KM, Robel P, Baulieu EE. Hydroxysteroid sulfotransferase activity in the rat brain and liver as a function of age and sex. Steroids 1997; 62:427-36. [PMID: 9178430 DOI: 10.1016/s0039-128x(97)00013-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The high concentrations of dehydroepiandrosterone sulfate and pregnenolone sulfate in the mammalian brain, despite the blood-brain barrier's impermeability to these compounds, and the apparent independence of these concentrations from those in plasma prompted us to investigate whether enzymatic sulfation of dehydroepiandrosterone was detectable in the rat brain. Low hydroxysteroid sulfotransferase activities were detectable in in vitro incubations of homogenates from all rat brain regions except the cerebellum, being highest in the hypothalamus and pons. This activity was not ascribable to enzyme in brain capillary blood. The activity was mainly cytosolic, although there was also significant activity in the partially purified nuclear fraction. The enzyme had different properties from those of hepatic isozymes, with a pH optimum of 6.5 and a high Km of approximately 2 mM for dehydroepiandrosterone. The enzyme was also active with pregnenolone as substrate. Activities in the brain were approximately 300-fold lower than in the liver but, as in the liver, these were higher in females than in males. The variations in brain activity as a function of age did not parallel those in the liver. Relatively high activities were found in the fetal brain and declined at birth, while activities were insignificant in the fetal liver and rose following birth. There was a major peak in activity in pubertal female brains, but this peak was less important, and later, in males. No evidence was found to indicate that the low brain enzyme activities and high Km were attributable either to the presence of an inhibitor or to the steroid sulfation actually being a secondary activity of another brain sulfotransferase. We discuss whether the sulfotransferase activities found are adequate to synthesize the dehydroepiandrosterone and pregnenolone sulfate found in brain.
Collapse
|
91
|
Bentvelsen FM, McPhaul MJ, Wilson JD, George FW. The androgen receptor of the urogenital tract of the fetal rat is regulated by androgen. Mol Cell Endocrinol 1994; 105:21-6. [PMID: 7821714 DOI: 10.1016/0303-7207(94)90031-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
To provide insight into androgen-mediated virilization, we measured the androgen receptor in tissues of male and female rat fetuses prior to and during the period of phenotypic sex differentiation. Western immunoblotting was performed utilizing an antibody directed against the 21 amino-terminal segment of the androgen receptor. In immunoblots prepared from urogenital tract tissues of day 17 male and female fetal rats, this antibody specifically recognizes a 110K protein band characteristic of the androgen receptor. Androgen receptor levels were low to undetectable in a variety of non-urogenital tract tissues. After day 18 of fetal development, the amount of androgen receptor decreased in female urogenital tissues, and by day 22 the amount of immunoreactive androgen receptor was higher in the male urogenital sinus and tubercle than in the corresponding tissues of the female. Administration of 5 alpha-dihydrotestosterone to pregnant rats at a dose of 50 mg/kg body weight per day from day 12 to day 22 caused an increase in immunoreactive androgen receptor in the female urogenital sinus and tubercle to levels approaching those in male tissues. Administration of the androgen antagonist flutamide (100 mg/kg body weight per day) during the same interval caused a reduction in androgen receptor level in the urogenital sinus and tubercle of the male. These findings suggest that androgens modulate the amount of androgen receptor in the embryonic urogenital tract either by inducing the proliferation of androgen-responsive cells or by increasing androgen receptor levels in individual cells.
Collapse
Affiliation(s)
- F M Bentvelsen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas 75235
| | | | | | | |
Collapse
|
92
|
Corbier P, Allioli N, Roffi J. [Variations in the testicular response to HCG during the perinatal period in the rat: influence of estrogens]. ARCHIVES INTERNATIONALES DE PHYSIOLOGIE, DE BIOCHIMIE ET DE BIOPHYSIQUE 1992; 100:389-97. [PMID: 1282394 DOI: 10.3109/13813459209000731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The variations of the testicular responsiveness to hCG and the implication of the maternal estrogens in the functioning of the testes were studied in the perinatal male rat. Male rat fetuses treated with hCG at the end of gestation failed to show an increase in serum testosterone (T). The lack of testicular responsiveness to hCG in the fetus is neither due to anesthesia nor to a blocking effect of estrogens directly on the testes. On the other hand, hCG injected either at 4 h or at 48 h after birth increases serum T. The administration of 5 micrograms of estradiol 17 beta (E2) to the newborn male rat at the time of birth blocks the expression of the postpartum testosterone surge. The fall in the plasma estrogens and the increase of the testicular sensitivity to gonadotropic stimulation at the time of birth are factors which are very likely implicated in the determinism of the neonatal testicular hyperactivity.
Collapse
Affiliation(s)
- P Corbier
- Laboratoire d'Endocrinologie, Université Paris XI, Centre d'Orsay, France
| | | | | |
Collapse
|
93
|
Habert R, Rouiller-Fabre V, Lecerf L, Levacher C, Saez JM. Developmental changes in testosterone production by the rat testis in vitro during late fetal life. ARCHIVES OF ANDROLOGY 1992; 29:191-7. [PMID: 1456840 DOI: 10.3109/01485019208987724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The age-related evolution of the in vitro capacity of the rat testis to produce testosterone and to respond to luteinizing hormone (LH) during 3 and 24 h incubation was studied from day 18.5 to day 21.5 of fetal life. Basal testosterone production by testes from 18.5-day-old fetuses was significantly higher than production by testes from 20.5- and 21.5-day-old fetuses when secretion was expressed as either per testis or per microgram of testicular protein. When maximal LH-stimulated testosterone secretion was expressed on a per testis basis, it was significantly lower for day 18.5 testes than for day 20.5 and 21.5 testes. However, when it was expressed on the basis of testicular protein content, it decreased significantly between days 18.5 and 20.5. Basal and LH-stimulated secretions displayed the same time-related decrease throughout 24 h of incubation for the three ages studied. The dose-response curves for LH showed that the sensitivity was similar for day 18.5 and 20.5 testes (ED50 = 10 vs. 14 ng/mL, respectively). These results showed an age-related decrease in testicular steroidogenic capacity without a change in the coupling efficiency of LH receptor to testosterone production during late fetal life in rats.
Collapse
Affiliation(s)
- R Habert
- Unité de Recherche sur la Différenciation et la Croissance Cellulaire INSERUM U 307, Université Paris
| | | | | | | | | |
Collapse
|
94
|
Habert R, Devif I, Gangnerau MN, Lecerf L. Ontogenesis of the in vitro response of rat testis to gonadotropin-releasing hormone. Mol Cell Endocrinol 1991; 82:199-206. [PMID: 1794610 DOI: 10.1016/0303-7207(91)90032-n] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The age-related evolution of the in vitro effects of a gonadotropin releasing hormone agonist ([D-Trp6]-GnRH) on the secretion of testosterone by the testis, cultured during 3 days on a Millipore filter floating on M199 medium, was studied during the perinatal period in the rat. The basal and luteinizing hormone (LH)-stimulated secretions by testes explanted on fetal day 14.5 were unaffected by the agonist. With fetal testes explanted on days 16.5 and 18.5 post-conception, the agonist inhibited, in a concentration-dependent manner, both basal and LH-stimulated secretions from the second or the third day of treatment onwards. With fetal and neonatal testes explanted on days 20.5, 21.5 and 31.5 post-conception, the GnRH agonist also had a long-term inhibitory effect on LH-stimulated secretion, but increased basal secretion. This stimulatory effect was already observed after 4 h of culture, and was maintained for 3 days. These results suggest that, during fetal development, the cellular mechanisms involved in the negative testicular response to GnRH are differentiated 3-5 days before those involved in the positive response. Lastly, after 3 days of preculture in hormone-free medium, fetal testes explanted on day 14.5 displayed long-term GnRH agonist inhibition of in vitro basal secretion of testosterone. This observation points out a spontaneous differentiation of the negative responsiveness to GnRH in the cultured fetal testis.
Collapse
Affiliation(s)
- R Habert
- Laboratoire de Physiologie du Developpement, CNRS URA 307 and INSERM U 307, Université Paris 7, France
| | | | | | | |
Collapse
|
95
|
Habert R, Brignaschi P. Developmental changes in in vitro testosterone production by dispersed Leydig cells during fetal life in rats. ARCHIVES OF ANDROLOGY 1991; 27:65-71. [PMID: 1953198 DOI: 10.3109/01485019108987654] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The age-related evolution during fetal days 18.5-21.5 of the capacity of collagenase-dispersed Leydig cells to produce testosterone and to respond to LH or dibutyryl cyclic AMP (dcAMP) was studied in vitro in the rat. When dispersed Leydig cells were incubated in control medium, testosterone secretion by cells from 18.5-day-old fetuses during the first 5 h was 100 and 50% higher than secretion by cells from 20.5- and 21.5-day-old fetuses, respectively. The secretion maximally stimulated by 100 ng/mL LH or stimulated by 1 mM dcAMP was also stronger on day 18.5 than on days 20.5 and 21.5 during the first 3 h of culture. The dose-response curves for LH showed that the ED50 was similar for day 18.5 and 20.5 cells (2 ng/mL LH). During the course of 24-h incubation, the secretion rates also changed with time and fetal age: Between 5 and 24 h of culture basal secretion decreased in day 18.5 cells, rose slightly in day 20.5 cells, and increased sharply in day 21.5 cells; in the same way, in the presence of LH or dcAMP, the secretion by day 18.5 cells decreased faster than that of day 20.5 or 21.5 cells. The basal testosterone secretion of the Leydig cells and its maximum steroidogenic capacity decrease during late fetal life in the rat, and there was no change in the sensitivity to LH during this period. The age-related differences in the variations of the secretion rates as a function of the duration of incubation suggest the existence of an evolution in extragonadotropic regulations during late fetal life.
Collapse
Affiliation(s)
- R Habert
- Laboratoire de Physiologie du Développement, University of Paris, France
| | | |
Collapse
|
96
|
Vega Matuszczyk J, Silverin B, Larsson K. Influence of environmental events immediately after birth on postnatal testosterone secretion and adult sexual behavior in the male rat. Horm Behav 1990; 24:450-8. [PMID: 2286361 DOI: 10.1016/0018-506x(90)90034-u] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A rise in plasma testosterone (T) levels occurs in male rats during the first 2 hr after birth which is of importance for the process of sexual differentiation. To study the influence of environmental factors on the postnatal T surge and sexual development, newborn male rats were subjected to various treatments immediately after cesarean delivery including cooling, ether anesthesia, and mother-infant separation. In adulthood, the animals were observed for masculine and feminine sexual behavior. Males anesthetized at 0 hr showed elevated levels of feminine sexual behavior and impaired masculine sexual behavior. Pups subjected to cooling or mother-infant separation showed slightly prolonged intromission latencies, but otherwise normal levels of feminine sexual behavior. Significantly elevated plasma T levels were found in intact pups 2 hr after birth but not in pups subjected to cooling or ether anesthesia. Significantly higher levels of T were observed in pups subjected to cooling 4 hr after birth, suggesting a delay of the T surge. The most pronounced impairing effects were seen in the defeminization process, but the masculinization process also is affected by ether anesthesia. It was concluded that ether anesthesia immediately after birth may permanently interfere with the sexual development by suppressing the neonatal T surge.
Collapse
|
97
|
Anderson CA, Clark RL. External genitalia of the rat: normal development and the histogenesis of 5 alpha-reductase inhibitor-induced abnormalities. TERATOLOGY 1990; 42:483-96. [PMID: 2177574 DOI: 10.1002/tera.1420420505] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The normal histogenesis of the rat genital tubercle and the effect of exposure in utero to the 5 alpha-reductase inhibitor finasteride (L-652,931; MK-0906; Proscar) on that process were studied. In normal males and females, the genital tubercle was first seen on Day 14.25 of gestation. It contained a urethral plate which extended from the cloaca (and after Day 15.25, from the urogenital sinus) to the tip of the tubercle. On Day 18.25 the glans lamellae, which would separate the glans penis or the clitoris from the prepuce, began to develop in both sexes. Also on Day 18.25 a dense, midline plate of mesenchymal cells was first evident between the urogenital sinus and the rectum in normal males. This plate acted as a wedge, first increasing the separation between the rectum and the urogenital sinus, and subsequently separating the urethral plate from the surface epithelium in the genital tubercle. As a result, by Day 21.25 the urethra in males followed an "S"-shaped course, extending from the pelvis through the center of the glans penis to an orifice near the tip of the genital tubercle. In females, in which a mesenchymal plate did not develop, the urethral orifice remained at the base of the tubercle, and the clitoris contained the remnants of the urethral plate, extending as an open groove from the urethral orifice to the tip of the tubercle. Finasteride did not affect development of the genital tubercle in females. However, in males exposed to finasteride in utero, there was variable failure of the mesenchymal wedge to develop. As a result, the urethral plate remained in contact with the surface epithelium and eventually opened to form a groove on the ventral surface of the glans penis (hypospadias). Also, the persistence of the urethral plate along the ventral midline in finasteride-treated male fetuses and its subsequent opening as a groove interfered with development of the glans lamellae, causing displacement of the frenulum distally on the glans penis and the development of a cleft in the prepuce.
Collapse
Affiliation(s)
- C A Anderson
- Department of Safety Assessment, Merck Sharp and Dohme Research Laboratories, West Point, Pennsylvania 19486
| | | |
Collapse
|
98
|
Clark RL, Antonello JM, Grossman SJ, Wise LD, Anderson C, Bagdon WJ, Prahalada S, MacDonald JS, Robertson RT. External genitalia abnormalities in male rats exposed in utero to finasteride, a 5 alpha-reductase inhibitor. TERATOLOGY 1990; 42:91-100. [PMID: 2168096 DOI: 10.1002/tera.1420420111] [Citation(s) in RCA: 96] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A series of studies was conducted to determine the developmental toxicity of the 5 alpha-reductase inhibitor finasteride (MK-0906) in rats. This compound was administered orally once daily to pregnant rats during various extended treatment periods during gestation. F1 offspring were evaluated on Day 20 of gestation as well as postnatally through mating to produce an F2 generation. MK-0906 treatment induced dosage-related incidences of hypospadias (penischisis) in male offspring with a threshold dosage level near 0.1 mg/kg/day and a 100% effect level of 100 mg/kg/day (with dosing through Day 20 of gestation). MK-0906 also caused decreased anogenital distance in male offspring. The dosage response for this effect (ranging from a 4.2% decrease at 0.003 mg/kg/day to a 38% decrease at 100 mg/kg/day) was more shallow than that for hypospadias. The decreases in anogenital distance were at least partially reversible postnatally with essentially complete recovery at dosages up to 0.1 mg/kg/day. There was also a dosage-related, temporary induction of nipples in F1 males. All of these effects were apparent following treatment on Days 6 through 17 of gestation but were more pronounced when dosing extended to Day 20 of gestation. Slight maternal toxicity consisting of minor decreases in body weight gain occurred only at dosages of 3 mg/kg/day and higher, indicating the selective nature of the developmental toxicity. The 5 alpha-reductase enzyme located in the rat fetal genital tubercle was studied in vitro and compared to that in the adult ventral prostate. The values for Km, Vmax, and IC50 for inhibition by MK-0906 were similar in the two tissues, suggesting that the enzymatic proteins in the genital tubercle and ventral prostate may be similar.
Collapse
Affiliation(s)
- R L Clark
- Merck Sharp & Dohme Research Laboratories, West Point, Pennsylvania 19486
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Dahlgren IL, Eriksson CJ, Gustafsson B, Harthon C, Hård E, Larsson K. Effects of chronic and acute ethanol treatment during prenatal and early postnatal ages on testosterone levels and sexual behaviors in rats. Pharmacol Biochem Behav 1989; 33:867-73. [PMID: 2616606 DOI: 10.1016/0091-3057(89)90484-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This study was prompted by previous findings that prenatal ethanol exposure may interfere with the differentiation of the sexual behavior in rats. Ethanol (6 g/kg) administered daily from day 15 postconception, resulted in elevated testosterone (T) levels on Day 18 in male and female fetuses. No alterations of sexual behavior in the ethanol-treated male offspring were seen under these conditions. However, in ethanol-treated female offspring the onset of regular estrous cycling was significantly delayed. Acute treatment with doses of ethanol, 2, 4 or 6 g/kg, was ineffective in influencing plasma T levels of the fetuses. Acute treatment with 3 g/kg ethanol did not prevent the rise of T levels normally occurring immediately after birth. In adulthood, but not at prepubertal age (Day 30), treatment of male rats with 2 g/kg ethanol caused a depression of plasma T levels. Possible mechanisms affected by ethanol exposure and influencing on the fetal development were discussed.
Collapse
Affiliation(s)
- I L Dahlgren
- Department of Psychology, University of Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
100
|
Habert R, Veniard B, Brignaschi P, Gangnerau MN, Picon R. Absence of development of late steroidogenic lesions in rat testis during the end of fetal life. ARCHIVES OF ANDROLOGY 1989; 22:41-8. [PMID: 2712640 DOI: 10.3109/01485018908986749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In the rat during the last 4 days of fetal life, the production of testosterone by the testis does not increase, whereas the plasma level of biological LH-like activity rises sharply. The present study was designed to test whether this phenomenon is attributed to the development of defects in the activity of the enzyme 17 alpha-hydroxylase and C17,20-lyase (called late steroidogenic lesions) during the end of fetal life. As this occurrence would lead to an age-related decrease in the ratio testosterone/progesterone, the testicular productions of these two steroids were evaluated on 18.5, 20.5, and 21.5 days postconception. Three different measurements were performed: (a) the in vivo testicular contents; (b) the in vitro secretions during a 120-min incubation in the presence or in the absence of 100 ng/ml ovine LH; (c) the testicular contents after these incubations. These measurements never revealed a decrease in the ratio testosterone/progesterone as a function of fetal age. It would appear that late steroidogenic lesions do not develop during the end of fetal life in the rat.
Collapse
Affiliation(s)
- R Habert
- Laboratoire de Physiologie du Développement et U.A. C.N.R.S. no. 307, Universite Paris, France
| | | | | | | | | |
Collapse
|