Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1293068, 2023.
Article in English | MEDLINE | ID: mdl-38304612

ABSTRACT

The stem cell niche plays a crucial role in the decision to either self-renew or differentiate. Recent observations lead to the hypothesis that O2 supply by blood and local O2 tension could be key components of the testicular niche of spermatogonial stem cells (SSCs). In this study, we investigated the impact of different hypoxic conditions (3.5%, 1%, and 0.1% O2 tension) on murine and human SSCs in culture. We observed a deleterious effect of severe hypoxia (1% O2 and 0.1% O2) on the capacity of murine SSCs to form germ cell clusters when plated at low density. Severe effects on SSCs proliferation occur at an O2 tension ≤1% and hypoxia was shown to induce a slight differentiation bias under 1% and 0.1% O2 conditions. Exposure to hypoxia did not appear to change the mitochondrial mass and the potential of membrane of mitochondria in SSCs, but induced the generation of mitochondrial ROS at 3.5% and 1% O2. In 3.5% O2 conditions, the capacity of SSCs to form colonies was maintained at the level of 21% O2 at low cell density, but it was impossible to amplify and maintain stem cell number in high cell density culture. In addition, we observed that 3.5% hypoxia did not improve the maintenance and propagation of human SSCs. Finally, our data tend to show that the transcription factors HIF-1α and HIF-2α are not involved in the SSCs cell autonomous response to hypoxia.

2.
Gynecol Obstet Fertil Senol ; 46(1): 41-46, 2018 Jan.
Article in French | MEDLINE | ID: mdl-29226802

ABSTRACT

Oogenesis is a complex cellular and molecular process whose fundamental mechanisms are still poorly described or not yet elucidated, especially in human species. The development of an in vitro model of oogenesis, particularly during fetal development in humans, is a critical step that would allow: (i) a better understand of the biological mechanisms of oogenesis; (ii) a refinement of medical diagnosis for women suffering from infertility; and (iii) providing new therapeutics for reproductive pathologies. The genesis of this model could be considered from ES/iPS cells. In this article, we will trace the physiological mechanisms of oogenesis in vivo and discuss the studies carried out in the field of in vitro oogenesis from ES/iPS cells, as well as the challenges to be met in the future.


Subject(s)
Oogenesis/physiology , Animals , Cell Differentiation , Fetal Development , Humans , Mice , Oogenesis/genetics , Pluripotent Stem Cells
3.
Reproduction ; 151(5): 477-89, 2016 May.
Article in English | MEDLINE | ID: mdl-26850882

ABSTRACT

Anti-silencing function 1 (ASF1) is an evolutionarily conserved histone H3-H4 chaperone involved in the assembly/disassembly of nucleosome and histone modification. Two paralogous genes, Asf1a and Asf1b, exist in the mouse genome. Asf1a is ubiquitously expressed and its loss causes embryonic lethality. Conversely, Asf1b expression is more restricted and has been less studied. To determine the in vivo function of Asf1b, we generated a Asf1b-deficient mouse line (Asf1b(GT(ROSA-ßgeo)437)) in which expression of the lacZ reporter gene is driven by the Asf1b promoter. Analysis of ß-galactosidase activity at early embryonic stages indicated a correlation between Asf1b expression and cell differentiation potential. In the gonads of both male and female, Asf1b expression was specifically detected in the germ cell lineage with a peak expression correlated with meiosis. The viability of Asf1b-null mice suggests that Asf1b is dispensable for mouse development. However, these mice showed reduced reproductive capacity compared with wild-type controls. We present evidence that the timing of meiotic entry and the subsequent gonad development are affected more severely in Asf1b-null female mice than in male mice. In female mice, in addition to subfertility related to altered gamete formation, variable defects compromising the development and/or survival of their offspring were also observed. Altogether, our data indicate the importance of Asf1b expression at the time of meiotic entry, suggesting that chromatin modifications may play a central role in this process.


Subject(s)
Cell Cycle Proteins/physiology , Chromosomal Proteins, Non-Histone/physiology , Fertility/genetics , Histones/metabolism , Nucleosomes/metabolism , Reproduction/physiology , Animals , Blotting, Western , Cells, Cultured , Female , Flow Cytometry , Histones/genetics , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleosomes/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
Cell Death Differ ; 23(3): 521-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26470726

ABSTRACT

Beside its well-documented role in carcinogenesis, the function of p53 family has been more recently revealed in development and female reproduction, but it is still poorly documented in male reproduction. We specifically tested this possibility by ablating Mdm2, an E3 ligase that regulates p53 protein stability and transactivation function, specifically in Sertoli cells (SCs) using the AMH-Cre line and created the new SC-Mdm2(-/-) line. Heterozygous SC-Mdm2(-/+) adult males were fertile, but SC-Mdm2(-/-) males were infertile and exhibited: a shorter ano-genital distance, an extra duct along the vas deferens that presents a uterus-like morphology, degenerated testes with no organized seminiferous tubules and a complete loss of differentiated germ cells. In adults, testosterone levels as well as StAR, P450c17 (Cyp17a1) and P450scc (Cyp11a1) mRNA levels decreased significantly, and both plasma LH and FSH levels increased. A detailed investigation of testicular development indicated that the phenotype arose during fetal life, with SC-Mdm2(-/-) testes being much smaller at birth. Interestingly, Leydig cells remained present until adulthood and fetal germ cells abnormally initiated meiosis. Inactivation of Mdm2 in SCs triggered p53 activation and apoptosis as early as 15.5 days post conception with significant increase in apoptotic SCs. Importantly, testis development occurred normally in SC-Mdm2(-/-) lacking p53 mice (SC-Mdm2(-/-)p53(-/-)) and accordingly, these mice were fertile indicating that the aforementioned phenotypes are entirely p53-dependent. These data not only highlight the importance of keeping p53 in check for proper testicular development and male fertility but also certify the critical role of SCs in the maintenance of meiotic repression.


Subject(s)
Apoptosis , Carrier Proteins/genetics , Infertility, Male/genetics , Sertoli Cells/physiology , Tumor Suppressor Protein p53/physiology , Animals , Carrier Proteins/metabolism , Gene Knockout Techniques , Infertility, Male/blood , Luteinizing Hormone/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Seminiferous Tubules/metabolism , Seminiferous Tubules/pathology , Testosterone/blood
5.
Cell Death Dis ; 4: e774, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23949223

ABSTRACT

Rad54 is an important factor in the homologous recombination pathway of DNA double-strand break repair. However, Rad54 knockout (KO) mice do not exhibit overt phenotypes at adulthood, even when exposed to radiation. In this study, we show that in Rad54 KO mouse the germline is actually altered. Compared with the wild-type (WT) animals, these mice have less premeiotic germ cells. This germ cell loss is found as early as in E11.5 embryos, suggesting an early failure during mutant primordial germ cells development. Both testicular and ovarian KO germ cells exhibited high radiation sensitivity leading to a long-term gametogenesis defect at adulthood. The KO female germline was particularly affected displaying decreased litter size or sterility. Spermatogenesis recovery after irradiation was slower and incomplete in Rad54 KO mice compared with that of WT mice, suggesting that loss of germ stem cell precursors is not fully compensated along the successive rounds of spermatogenesis. Finally, spermatogenesis recovery after postnatal irradiation is in part regulated by glial-cell-line-derived neurotrophic factor (GDNF) in KO but not in irradiated WT mice, suggesting that Sertoli cell GDNF production is stimulated upon substantial germ cell loss only. Our findings suggest that Rad54 has a key function in maintaining genomic integrity of the developing germ cells.


Subject(s)
DNA Damage , DNA Helicases/metabolism , Genomic Instability , Germ Cells/pathology , Nuclear Proteins/metabolism , Animals , Cell Count , Cell Death/genetics , Cell Death/radiation effects , DNA Damage/radiation effects , DNA Helicases/deficiency , Dose-Response Relationship, Radiation , Female , Fetus/metabolism , Fetus/radiation effects , Gamma Rays , Genomic Instability/radiation effects , Germ Cells/metabolism , Germ Cells/radiation effects , Infertility, Female/embryology , Infertility, Female/pathology , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Ovary/embryology , Ovary/pathology , Ovary/radiation effects , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Sertoli Cells/pathology , Spermatogenesis/genetics , Spermatogenesis/radiation effects , Spermatogonia/metabolism , Spermatogonia/pathology , Spermatogonia/radiation effects , Testis/embryology , Testis/pathology , Testis/radiation effects
6.
Hum Reprod ; 25(10): 2579-90, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20670969

ABSTRACT

BACKGROUND: The initiation of meiosis is crucial to fertility. While extensive studies in rodents have enhanced our understanding of this process, studies in human fetal ovary are lacking. METHODS: We used RT-PCR and immunohistochemistry to investigate expression of meiotic factors in human fetal ovaries from 6 to 15 weeks post fertilization (wpf) and developed an organ culture model to study the initiation of human meiosis. RESULTS: We observed the first meiotic cells at 11 wpf, when STRA8, SPO11 and DMC1 are first expressed. In culture, meiosis initiation is observed in 10 and 11 wpf ovaries and meiosis is maintained by addition of fetal calf serum. Meiosis is stimulated, compared with control, by retinoic acid (RA) (P < 0.05). No major change occurred in mRNA for CYP26B1, the RA-degrading enzyme proposed to control the timing of meiosis in mice. We did, however, observe increased mRNA levels for ALDH1A1 in human ovary when meiosis began, and evidence for a requirement to synthesize RA and thus sustain meiosis. Indeed, ALDH inhibition by citral prevented the appearance of meiotic cells. Finally, 8 wpf ovaries (and earlier stages) were unable to initiate meiosis whatever the length of culture, even in the presence of RA and serum. However, when human germ cells from 8 wpf ovaries were placed in a mouse ovarian environment, some did initiate meiosis. CONCLUSIONS: Our data indicate that meiosis initiation in the human ovary relies partially on RA, but that the progression and regulation of this process appears to differ in many aspects from that described in mice.


Subject(s)
Meiosis , Ovary/cytology , Tretinoin/metabolism , Acyclic Monoterpenes , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Cytochrome P-450 Enzyme System/metabolism , Female , Fetus , Humans , Mice , Monoterpenes/pharmacology , Ovary/drug effects , Ovary/metabolism , Ovum/cytology , Ovum/drug effects , Ovum/metabolism , Retinal Dehydrogenase , Retinoic Acid 4-Hydroxylase , Tretinoin/pharmacology
7.
Cell Death Differ ; 14(4): 671-81, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17082817

ABSTRACT

In mammals, the pool of primordial follicles at birth is determinant for female fertility. Exposure to IR during oogonia proliferation and the diplotene stages of ovarian development induced the virtual disappearance of primordial follicles in the postnatal ovary, while half the follicular reserve remained present after irradiation during the zygotene/pachytene stages. This sensitivity difference was correlated with the level of caspase-2 expression evaluated by immunohistochemistry. At the diplotene stage, Western blot and caspase activity analysis revealed that caspase-2 was activated 2 h after irradiation and a significant increase in the number of oocytes expressing cleaved caspase-9 and -3 occurred 6 h after treatment. Inhibition of caspase-2 activity prevented the cleavage of caspase-9 and partially prevented the loss of oocytes in response to irradiation. Taken together, our results show that caspase-2-dependent activation of the mitochondrial apoptotic pathway is one of the mechanisms involved in the genotoxic stress-induced depletion of the primordial follicle pool.


Subject(s)
Apoptosis/radiation effects , Caspase 2/metabolism , Fetus/radiation effects , Oocytes/radiation effects , Ovary/radiation effects , Animals , Apoptosis/physiology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Fertility/radiation effects , Fetus/enzymology , Gene Regulatory Networks/genetics , Gene Regulatory Networks/radiation effects , Humans , Immunohistochemistry , Mice , Mice, Inbred Strains , Oocytes/enzymology , Ovary/embryology , Ovary/enzymology , Pregnancy
8.
Int J Radiat Biol ; 82(11): 771-80, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17148261

ABSTRACT

PURPOSE: To investigate the role of p63, a member of the p53 family, in gonocyte apoptosis after radiation exposure. MATERIALS AND METHODS: Wild-type (WT) and p63 knock-out (KO) testes were exposed in vivo or in vitro to a 3 Gy dose of 137Cesium (137Cs) gamma-rays at day 18.5 post-conception (p.c.). p63 whole expression was studied in neonatal testes by immunohistochemistry, whereas TAp63 and DeltaNp63 isoforms were studied by Reverse-transcribed Polymerase Chain Reaction (RT-PCR). Gonocyte apoptosis was analysed by immunohistochemistry (cleaved caspase 3) and In Situ End labelling (ISEL). RESULTS: Such foetal irradiation leads to a strong increase of gonocyte apoptosis in newborns. It also induces the up-regulation of the TAp63alpha isoform and the down-regulation of the DeltaNp63alpha isoform. Moreover, in control p63KO testis, a significant increase in the number of gonocytes was associated with a strong reduction of their apoptosis compared with the control wild-type testis. Unexpectedly, after irradiation this increase of the number of apoptotic gonocytes was seen in p63KO testis, which was comparable to that in irradiated p63WT testis. CONCLUSION: We demonstrate that p63 is able to trigger gonocyte apoptosis in control testis but is not necessarily required in their radio-induced apoptosis.


Subject(s)
Apoptosis/radiation effects , Fetus/radiation effects , Phosphoproteins/physiology , Spermatozoa/radiation effects , Trans-Activators/physiology , Animals , Animals, Newborn , Cell Survival/radiation effects , Immunohistochemistry , Male , Mice , Mice, Knockout , Phosphoproteins/analysis , Radiation Tolerance , Spermatozoa/pathology , Testis/chemistry , Testis/radiation effects , Trans-Activators/analysis , Tumor Suppressor Protein p53/physiology
9.
Biochimie ; 88(11): 1831-5, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17070978

ABSTRACT

Exposure to environmental pollutants (EP) is associated with a wide range of toxic effects, in particular in testis development. Uranium is a potential pollutant of nuclear industry and over the last few years, its environmental concentrations have increased. In animals, the current procedures for evaluating the potential developmental toxicity of uranium are based on in vivo studies. These methods do not allow to know the direct effects on testicular cells and are obviously excluded for human experiments. Consequently, we have developed an in vitro culture system of the whole testis. In the present study we characterized and validated this organ culture system in both mouse fetal testes and human fetal testes recovered during the first trimester (6-12 weeks) of gestation. We compared the histological aspect, the number of germ cells and the testosterone production, before and after culture. Testicular architecture and intercellular communications were preserved, and organ culture appears as a powerful method for studying the early development of testicular gametogenesis and steroidogenesis in both species. Thus by using this method we will be able to investigate the effects of uranium on mouse and human developing testis. The mouse model will allow us to determine the dose range of interest without restriction of material.


Subject(s)
Environmental Pollutants/toxicity , Testis/embryology , Animals , Fetal Development , Humans , Male , Mice , Mice, Inbred Strains , Organ Culture Techniques , Testis/drug effects , Testis/pathology
10.
Gynecol Obstet Fertil ; 34(10): 970-7, 2006 Oct.
Article in French | MEDLINE | ID: mdl-16971153

ABSTRACT

Estrogens are classically known to play a major role in female reproduction but there is now compelling evidence that they may also be involved in the regulation of male reproductive function. In humans, a decrease in sperm count and an increase in the incidences of testicular cancer, cryptorchidism and hypospadia have been observed in many countries over the last 50 years. Male reproductive alterations were also observed in wildlife. Such male reproductive disorders have been attributed to the increase in concentration of xenobiotics, and of xenoestrogens in particular, in the environment and in food. Epidemiological, clinical and experimental studies have suggested that excessive exposure to estrogens during fetal/neonatal life can lead to reproductive disorders in adulthood. Using an in vitro model we showed that estrogens directly affected the development of the fetal testis and we evidenced the existence of periods of sensitivity throughout development. Lastly, we clearly demonstrated that the fetal and neonatal testis is very sensitive to estrogens since the invalidation of estrogen receptor alpha leads to an increase of steroidogenesis and the invalidation of estrogen receptor beta enhances the development of the germ cell lineage in the male.


Subject(s)
Estrogens/physiology , Testis/embryology , Testis/growth & development , Cryptorchidism/epidemiology , Estrogens/administration & dosage , Estrogens, Non-Steroidal , Female , Humans , Hypospadias/epidemiology , Infant, Newborn , Male , Pregnancy , Receptors, Estrogen/physiology , Sperm Count , Testicular Neoplasms/epidemiology , Testis/drug effects , Xenobiotics
11.
Mol Endocrinol ; 19(5): 1277-90, 2005 May.
Article in English | MEDLINE | ID: mdl-15705663

ABSTRACT

Mammalian spermatids and spermatozoa express functional G protein-coupled receptors. However, bicarbonate-regulated soluble adenylyl cyclase (AC), the major AC present in these cells, is not directly coupled to G proteins. To understand how G protein-coupled receptors signal in spermatozoa, we investigated whether a conventional transmembrane cyclase is present and biologically active in these cells. Here, we provide evidence for expression of type 3 AC (AC3) in male germ cells and describe the effects of disruption of the AC3 gene on fertility and function of mouse spermatozoa. As previously reported in rat, AC3 mRNA is expressed in mouse testes and localized, together with soluble AC mRNA, mainly in postmeiotic germ cells. AC3 protein was detected by immunolocalization in round and elongating spermatids in a region corresponding to the developing acrosome and was retained in the mature spermatozoa of the epididymis. Forskolin caused a small increase in cAMP production in mouse spermatozoa, but this increase could not be detected in the AC3(-/-) mice. Inactivation of the AC3 gene did not have overt effects on spermatogenesis; however, AC3(-/-) males were subfertile with only three litters generated by 11 males over a period of 6 months. When used in in vitro fertilization, spermatozoa from these AC3(-/-) mice produced few embryos, but their fertilizing ability was restored after removal of the zona pellucida. Despite an apparently normal structure, these spermatozoa had decreased motility and showed an increase in spontaneous acrosome reactions. These data support the hypothesis that AC3 is required for normal spermatid or spermatozoa function and male fertility.


Subject(s)
Adenylyl Cyclases/genetics , Infertility, Male/genetics , Isoenzymes/genetics , Spermatozoa/metabolism , Adenylyl Cyclases/biosynthesis , Animals , Cyclic AMP/metabolism , Epididymis/abnormalities , Fertilization/genetics , Immunohistochemistry , Infertility, Male/metabolism , Isoenzymes/biosynthesis , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , Sequence Analysis, DNA , Sperm Motility/genetics , Testis/abnormalities , Testis/metabolism
12.
Biol Reprod ; 70(6): 1814-21, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14960491

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.


Subject(s)
Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis , Tretinoin/pharmacology , Animals , Animals, Newborn , Base Sequence , Cell Differentiation/drug effects , DNA/genetics , Female , Fetus/drug effects , Fetus/metabolism , Gestational Age , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Male , Organ Culture Techniques , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/genetics , Testis/cytology , Vitamin A Deficiency/metabolism
13.
Andrologia ; 35(1): 79-83, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558532

ABSTRACT

The foetal testis originates from a proliferation of the mesonephric and the coelomic epithelia which are colonized by the primordial germ cells. In the foetal testis, the development and functions of the three main cell type precursors (Leydig, Sertoli and germ cells) do not depend upon gonadotropins. Numerous intra- and extra-testicular factors are candidates for the control of its development and functions. To study the potential involvement of these factors, we developed an organotypic culture system. In absence of any growth factors or hormone, this system allows a development of the three main cell types which mimics that observed in vivo. The effects of different regulators (gonadotropin-releasing hormone, follicle-stimulating hormone, transforming growth factor-beta, insulin-like growth factor-I, anti-Mullerian hormone, retinoic acid, oestrogens) were tested in this system. Whether or not some of the effects observed in vitro have a physiological relevance was evaluated using appropriate transgenic mice. It is concluded that the foetal testis cannot be considered as an adult mini-testis since it has a specific physiology which largely differs from that of the immature or adult testis.


Subject(s)
Testis/embryology , Testis/growth & development , Animals , Animals, Newborn/growth & development , Cell Differentiation , Gonadotropins/physiology , Humans , Male , Organ Culture Techniques
14.
Biol Reprod ; 64(5): 1307-14, 2001 May.
Article in English | MEDLINE | ID: mdl-11319133

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.


Subject(s)
Receptors, Retinoic Acid/physiology , Testis/embryology , Testis/growth & development , Tretinoin/pharmacology , Animals , Animals, Newborn , Cell Division/drug effects , Culture Techniques , Cyclic AMP/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Luteinizing Hormone/pharmacology , Male , Morphogenesis/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoid X Receptors , Retinoids/pharmacology , Sertoli Cells/drug effects , Testis/metabolism , Testosterone/biosynthesis , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology
15.
Mol Cell Endocrinol ; 172(1-2): 193-202, 2001 Feb 14.
Article in English | MEDLINE | ID: mdl-11165053

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.


Subject(s)
Cell Differentiation/drug effects , Leydig Cells/drug effects , Luteinizing Hormone/pharmacology , Animals , Cell Division/drug effects , Female , Fetus/cytology , Gestational Age , Leydig Cells/cytology , Male , Rats , Rats, Wistar , Testis/drug effects , Testis/embryology , Testis/growth & development , Testosterone/metabolism , Wolffian Ducts/drug effects , Wolffian Ducts/embryology , Wolffian Ducts/growth & development
16.
Mol Cell Endocrinol ; 165(1-2): 225-31, 2000 Jul 25.
Article in English | MEDLINE | ID: mdl-10940501

ABSTRACT

We investigated the effect of retinoids on the entrance of female germ cells into meiotic prophase and their progression through it, using explants of rat ovaries from 14.5 days post coïtum (dpc) fetuses cultured with or without 10(-6) M retinoic acid (RA) or 10(-9) M retinoic acid receptor alpha (RARalpha) specific agonist. The percentages of oogonia and of oocytes at each meiotic stage in the ovary were evaluated at explantation (D0) and after 3 (D3), 5 (D5) and 9 (D9) days of culture and on equivalent stages in vivo (i. e. 17.5 and 23.5 dpc). The number of germ cells per ovary were counted at D0, D3 and D9. Newly explanted (D0) ovaries contained no germ cell in meiosis. In control medium some germ cells had spontaneously reached the stage leptotene and very few the zygotene on D3. The first pachytene were observed on D5 and the first diplotene on D9. This pattern mimicked that which occurs in vivo although with a slight delay. RA reduced the percentage of oogonia by more than half and increased the percentage of zygotene by more than 22-fold on D3, showing that it accelerated entrance into meiosis. This effect was also observed in response to RARalpha agonist. RA increased the percentage of zygotene and reduced the percentage of pachytene on D9, showing that it can also delay the zygotene/pachytene transition. Lastly, RA reduced the total number of germ cells present on D3 but not on D9. This may be the result of a double effect of RA on the number of germ cells: negative when the cells are in proliferation (D0 to D3) and positive when they entered in meiotic prophase (after D3). Thus, RA is a potential regulator of germ cells meiosis and number in the fetal ovary.


Subject(s)
Meiosis/drug effects , Ovary/cytology , Ovary/drug effects , Retinoids/pharmacology , Animals , Benzoates/pharmacology , Female , Oogenesis/drug effects , Organ Culture Techniques , Ovary/embryology , Rats , Receptors, Retinoic Acid/agonists , Retinoic Acid Receptor alpha , Tetrahydronaphthalenes/pharmacology , Tretinoin/pharmacology
17.
Biol Reprod ; 62(5): 1303-14, 2000 May.
Article in English | MEDLINE | ID: mdl-10775181

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.


Subject(s)
Leydig Cells/drug effects , Retinoids/pharmacology , Sertoli Cells/drug effects , Testis/cytology , Testis/embryology , Animals , Animals, Newborn , Cell Division/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Spermatozoa/drug effects , Testis/drug effects , Testis/growth & development , Testosterone/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...