Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
DNA Repair (Amst) ; 102: 103097, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33812231

RESUMEN

During meiosis, programmed double-strand breaks are repaired by homologous recombination (HR) to form crossovers that are essential to homologous chromosome segregation. Single-stranded DNA (ssDNA) containing intermediates are key features of HR, which must be highly regulated. RPA, the ubiquitous ssDNA binding complex, was thought to play similar roles during mitotic and meiotic HR until the recent discovery of MEIOB and its partner, SPATA22, two essential meiosis-specific proteins. Here, we show that like MEIOB, SPATA22 resembles RPA subunits and binds ssDNA. We studied the physical and functional interactions existing between MEIOB, SPATA22, and RPA, and show that MEIOB and SPATA22 interact with the preformed RPA complex through their interacting domain and condense RPA-coated ssDNA in vitro. In meiotic cells, we show that MEIOB and SPATA22 modify the immunodetection of the two large subunits of RPA. Given these results, we propose that MEIOB-SPATA22 and RPA form a functional ssDNA-interacting complex to satisfy meiotic HR requirements by providing specific properties to the ssDNA.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Emparejamiento Cromosómico , Intercambio Genético , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína de Replicación A/metabolismo , Animales , Línea Celular Tumoral , Células HEK293 , Recombinación Homóloga , Humanos , Meiosis , Ratones , Modelos Moleculares , Complejos Multiproteicos , Conformación Proteica
2.
Hum Reprod ; 32(3): 631-642, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28073973

RESUMEN

STUDY QUESTION: How can pre-meiotic germ cells persist in the human foetal ovary? SUMMARY ANSWER: Numerous oogonia escaping meiotic entry were retrieved throughout human ovarian development simultaneously with the expression of signalling pathways preventing meiosis, typically described in the rodent embryonic testis. WHAT IS KNOWN ALREADY: The transition from mitosis to meiosis is a key event in female germ cells that remains poorly documented in research on the human ovary. Previous reports described a strikingly asynchronous differentiation in the human female germ line during development, with the persistence of oogonia among oocytes and follicles during the second and third trimesters. The possible mechanisms allowing some cells to escape meiosis remain elusive. STUDY DESIGN SIZE, DURATION: In order to document the extent of this phenomenon, we detailed the expression profile of germ cell differentiation markers using 73 ovaries ranging from 6.4 to 35 weeks post-fertilization. PARTICIPANTS/MATERIALS SETTING, METHODS: Pre-meiotic markers were detected by immunohistochemistry or qRT-PCR. The expression of the main meiosis-preventing factors identified in mice was analysed, and their functionality assessed using organ cultures. MAIN RESULTS AND THE ROLE OF CHANCE: Oogonia stained for AP2γ could be traced from the first trimester until the end of the third trimester. Female germ cell differentiation is organized both in time and space in a centripetal manner in the foetal human ovary. Unexpectedly, some features usually ascribed to rodent pre-spermatogonia could be observed in human foetal ovaries, such as NANOS2 expression and quiescence in some germ cells. The two main somatic signals known to inhibit meiosis in the mouse embryonic testis, CYP26B1 and FGF9, were detected in the human ovary and act simultaneously to repress STRA8 and meiosis in human foetal female germ cells. LARGE SCALE DATA: N/A. LIMITATIONS REASON FOR CAUTION: Our conclusions relied partly on in vitro experiments. Germ cells were not systematically identified with immunostaining and some may have thus escaped analysis. WIDER IMPLICATIONS OF THE FINDINGS: We found evidence that a robust repression of meiotic entry is taking place in the human foetal ovary, possibly explaining the exceptional long-lasting presence of pre-meiotic germ cells until late gestational age. This result calls for a redefinition of the markers known as classical male markers, which may in fact characterize mammalian developing gonads irrespectively of their sex. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by the Université Paris Diderot-Paris 7 and Université Paris-Sud, CEA, INSERM, and Agence de la Biomédecine. The authors declare no conflict of interest.


Asunto(s)
Células Germinales Embrionarias/metabolismo , Meiosis/fisiología , Ovario/embriología , Testículo/embriología , Animales , Proliferación Celular/fisiología , Femenino , Humanos , Masculino , Ratones , Oogonios/citología , Oogonios/metabolismo , Ovario/metabolismo , Transducción de Señal/fisiología , Espermatogonias/citología , Espermatogonias/metabolismo , Testículo/metabolismo
3.
FEBS Lett ; 590(16): 2566-74, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27364741

RESUMEN

Murine double minute 2 and 4 (Mdm2, Mdm4) are major p53-negative regulators, preventing thus uncontrolled apoptosis induction in numerous cell types, although their function in the female germ line has received little attention. In the present work, we have generated mice with specific invalidation of Mdm2 and Mdm4 genes in the mouse oocyte (Mdm2(Ocko) and Mdm4(Ocko) mice), to test their implication in survival of these germ cells. Most of the Mdm2(Ocko) but not Mdm4(Ocko) mice were sterile, with a dramatic reduction of the weight of ovaries and genital tract, a strong increase in follicle-stimulating hormone and luteinizing hormone serum levels, and a reduction of anti-mullerian hormone serum levels. Histological analyses revealed an obvious decrease of the number of growing follicles beyond the primary stage in Mdm2(Ocko) ovaries in comparison to controls, with a pronounced increase in the apparition of primary atretic follicles, most being devoid of oocyte. Similar phenotypes were observed with Mdm2(Ocko) Mdm4(Ocko) ovaries, with no worsening of the phenotype. However, we failed to detect any increase in p53 level in mutant oocytes, nor any other apoptotic marker, introgression of this targeted invalidation in p53-/- mice restored the fertility of females. This study is the first to show that Mdm2, but not Mdm4, has a critical role in oocyte survival and would be involved in premature ovarian insufficiency phenotype.


Asunto(s)
Infertilidad Femenina/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Hormona Antimülleriana/metabolismo , Femenino , Hormona Folículo Estimulante , Infertilidad Femenina/patología , Ratones , Ratones Noqueados , Oocitos/metabolismo , Oocitos/patología , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Ovario/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
PLoS Genet ; 9(9): e1003784, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24068956

RESUMEN

Meiotic recombination is a mandatory process for sexual reproduction. We identified a protein specifically implicated in meiotic homologous recombination that we named: meiosis specific with OB domain (MEIOB). This protein is conserved among metazoan species and contains single-strand DNA binding sites similar to those of RPA1. Our studies in vitro revealed that both recombinant and endogenous MEIOB can be retained on single-strand DNA. Those in vivo demonstrated the specific expression of Meiob in early meiotic germ cells and the co-localization of MEIOB protein with RPA on chromosome axes. MEIOB localization in Dmc1 (-/-) spermatocytes indicated that it accumulates on resected DNA. Homologous Meiob deletion in mice caused infertility in both sexes, due to a meiotic arrest at a zygotene/pachytene-like stage. DNA double strand break repair and homologous chromosome synapsis were impaired in Meiob (-/-) meiocytes. Interestingly MEIOB appeared to be dispensable for the initial loading of recombinases but was required to maintain a proper number of RAD51 and DMC1 foci beyond the zygotene stage. In light of these findings, we propose that RPA and this new single-strand DNA binding protein MEIOB, are essential to ensure the proper stabilization of recombinases which is required for successful homology search and meiotic recombination.


Asunto(s)
Emparejamiento Cromosómico/genética , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Recombinación Homóloga/genética , Meiosis/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Células Germinativas , Humanos , Masculino , Ratones , Recombinasa Rad51/genética , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Espermatocitos/metabolismo
5.
J Clin Endocrinol Metab ; 97(10): E1890-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22802086

RESUMEN

CONTEXT: The 21-hydroxylase deficiency is the most common cause of congenital adrenal hyperplasia. Pregnant women presenting a risk of genetic transmission may be treated with synthetic glucocorticoids such as dexamethasone (DEX) to prevent female fetus virilization. OBJECTIVE: The aim of this study was to assess the potential deleterious effects of DEX exposure on fetal ovarian development. SETTINGS: Human fetal ovaries, ranging from 8-11 weeks after fertilization, were harvested from material available after legally induced abortions. They were cultured in the absence or presence of DEX (2, 10, or 50 µm) over 14 d, and histological analyses were performed. RESULTS: The glucocorticoid receptor NR3C1 was present and the signaling pathway active in the fetal ovary as demonstrated by the expression of NR3C1 target genes, such as PLZF and FKBP5, in response to DEX exposure. DEX decreased germ cell density at the 10 and 50 µm doses. Exposure to DEX, even at the highest dose, did not change oogonial proliferation as monitored by 5-bromo-2'-deoxyuridine incorporation and significantly increased the apoptotic rate, detected with cleaved caspase 3 staining. Interestingly, the expression of the prosurvival gene KIT was significantly decreased in the presence of DEX during the course of the culture. CONCLUSION: We have demonstrated for the first time that in vitro exposure to high doses of DEX impairs human fetal oogenesis through an increase in apoptosis. These data are of high importance, and additional epidemiological studies are required to investigate the female fertility of those women who have been exposed to DEX during fetal life.


Asunto(s)
Hiperplasia Suprarrenal Congénita/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Dexametasona/efectos adversos , Oogonios/efectos de los fármacos , Ovario/citología , Ovario/efectos de los fármacos , Recuento de Células , Dexametasona/administración & dosificación , Femenino , Feto/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucocorticoides/administración & dosificación , Glucocorticoides/efectos adversos , Humanos , Oogonios/citología , Técnicas de Cultivo de Órganos , Embarazo , Proteínas Proto-Oncogénicas c-kit/genética , Receptores de Glucocorticoides/genética , Virilismo/prevención & control
6.
Development ; 138(24): 5393-402, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22071108

RESUMEN

The mechanisms regulating germ line sex determination and meiosis initiation are poorly understood. Here, we provide evidence for the involvement of homeobox Msx transcription factors in foetal meiosis initiation in mammalian germ cells. Upon meiosis initiation, Msx1 and Msx2 genes are strongly expressed in the foetal ovary, possibly stimulated by soluble factors found there: bone morphogenetic proteins Bmp2 and Bmp4, and retinoic acid. Analysis of Msx1/Msx2 double mutant embryos revealed a majority of undifferentiated germ cells remaining in the ovary and, importantly, a decrease in the number of meiotic cells. In vivo, the Msx1/Msx2 double-null mutation prevented full activation of Stra8, a gene required for meiosis. In F9 cells, Msx1 can bind to Stra8 regulatory sequences and Msx1 overexpression stimulates Stra8 transcription. Collectively, our data demonstrate for the first time that some homeobox genes are required for meiosis initiation in the female germ line.


Asunto(s)
Proteínas de Homeodominio/fisiología , Factor de Transcripción MSX1/fisiología , Meiosis/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Ratones , Mutación , Técnicas de Cultivo de Órganos , Ovario/fisiología , Proteínas/metabolismo , Tretinoina/metabolismo , Tretinoina/fisiología
7.
Reprod Toxicol ; 31(4): 470-6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21262344

RESUMEN

Uranium (U) is found in the environment and its use in industrial or military activities has led to concerns about its potential toxicity. The reprotoxicity of this heavy metal has been established in adult animals; however, no studies have examined its effect on human fetal gonads. Using an organ culture system, we investigated the effects of uranyl acetate on human gonads during the first trimester of gestation (7-12 weeks), which is a critical step in the development of a functional reproductive system. In human fetal ovaries, 0.05 mM U significantly decreased germ cell density by increasing their apoptosis rate. In human fetal testes, 0.1mM U similarly reduced the number of germ cells. The human fetal germ cells are more sensitive to U than mouse germ cells in the same experimental conditions. This is the first evidence that U may impair the development of the human gonads.


Asunto(s)
Compuestos Organometálicos/toxicidad , Ovario/efectos de los fármacos , Óvulo/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Recuento de Células , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Edad Gestacional , Humanos , Inmunohistoquímica , Masculino , Ratones , Técnicas de Cultivo de Órganos , Ovario/embriología , Ovario/patología , Óvulo/patología , Espermatozoides/patología , Testículo/embriología , Testículo/patología
8.
Dev Biol ; 346(2): 320-30, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20707993

RESUMEN

In mammals, early fetal germ cells are unique in their ability to initiate the spermatogenesis or oogenesis programs dependent of their somatic environment. In mice, female germ cells enter into meiosis at 13.5 dpc whereas in the male, germ cells undergo mitotic arrest. Recent findings indicate that Cyp26b1, a RA-degrading enzyme, is a key factor preventing initiation of meiosis in the fetal testis. Here, we report evidence for additional testicular pathways involved in the prevention of fetal meiosis. Using a co-culture model in which an undifferentiated XX gonad is cultured with a fetal or neonatal testis, we demonstrated that the testis prevented the initiation of meiosis and induced male germ cell differentiation in the XX gonad. This testicular effect disappeared when male meiosis starts in the neonatal testis and was not directly due to Cyp26b1 expression. Moreover, neither RA nor ketoconazole, an inhibitor of Cyp26b1, completely prevented testicular inhibition of meiosis in co-cultured ovary. We found that secreted factor(s), with molecular weight greater than 10 kDa contained in conditioned media from cultured fetal testes, inhibited meiosis in the XX gonad. Lastly, although both Sertoli and interstitial cells inhibited meiosis in XX germ cells, only interstitial cells induced mitotic arrest in germ cell. In conclusion, our results demonstrate that male germ cell determination is supported by additional non-retinoid secreted factors inhibiting both meiosis and mitosis and produced by the testicular somatic cells during fetal and neonatal life.


Asunto(s)
Meiosis , Testículo/embriología , Animales , Diferenciación Celular , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Mitosis , Ácido Retinoico 4-Hidroxilasa , Testículo/citología , Testículo/metabolismo
9.
Environ Health Perspect ; 118(3): 331-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20064782

RESUMEN

BACKGROUND: Cadmium (Cd) is a common environmental pollutant and a major constituent of tobacco smoke. Adverse effects of this heavy metal on reproductive function have been identified in adults; however, no studies have examined its effects on human reproductive organs during development. OBJECTIVES: Using our previously developed organ culture system, we investigated the effects of cadmium chloride on human gonads at the beginning of fetal life, a critical stage in the development of reproductive function. METHODS: Human fetal gonads were recovered during the first trimester (711 weeks postconception) and cultured with or without Cd. We used different concentrations of Cd and compared results with those obtained with mouse fetal gonads at similar stages. RESULTS: Cd, at concentrations as low as 1 microM, significantly decreased the germ cell density in human fetal ovaries. This correlated with an increase in germ cell apoptosis, but there was no effect on proliferation. Similarly, in the human fetal testis, Cd (1 microM) reduced germ cell number without affecting testosterone secretion. In mouse fetal gonads, Cd increased only female germ cell apoptosis. CONCLUSIONS: This is the first experimental demonstration that Cd, at low concentrations, alters the survival of male and female germ cells in humans. Considering data demonstrating extensive human exposure, we believe that current environmental levels of Cd could be deleterious to early gametogenesis.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Apoptosis/efectos de los fármacos , Cadmio/toxicidad , Feto/citología , Células Germinativas/efectos de los fármacos , Exposición Materna/efectos adversos , Adulto , Animales , Femenino , Feto/anatomía & histología , Células Germinativas/citología , Gónadas/citología , Gónadas/efectos de los fármacos , Gónadas/embriología , Humanos , Masculino , Ratones , Técnicas de Cultivo de Órganos , Embarazo , Primer Trimestre del Embarazo
10.
Hum Reprod ; 24(3): 670-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19088112

RESUMEN

BACKGROUND: We have previously shown that male human fetal germ cells are highly radiosensitive and that their death depends on p53 activation. Male germ cell apoptosis was initiated with doses as low as 0.1 Gy and was prevented by pifithrin alpha, a p53 inhibitor. In this study, we investigated the radiosensitivity of early female and male fetal proliferating germ cells. METHODS AND RESULTS: Both male and female fetal germ cells displayed a similar number of gamma H2AX foci in response to ionizing radiation (IR). In organ culture of human fetal ovaries, the germ cells underwent apoptosis only when exposed to high doses of IR (1.5 Gy and above). Accumulation of p53 was detected in irradiated male human fetal germ cells but not in female ones. Inhibition of p53 with pifithrin alpha did not affect oogonia apoptosis following irradiation. IR induced apoptosis similarly in mouse fetal ovaries in organ culture and in vivo during oogonial proliferation. Germ cell survival in testes from p53 knockout or p63 knockout mice exposed to IR was better than wild-type, whereas female germ cell survival was unaffected by p53 or p63 knockout. CONCLUSIONS: These findings show that pre-meiotic male and female fetal germ cells behave differently in response to a genotoxic stress--irradiation--with oogonia being less sensitive and undergoing p53-independent apoptosis.


Asunto(s)
Apoptosis , Células Germinativas/citología , Células Germinativas/efectos de la radiación , Factores Sexuales , Animales , Benzotiazoles/farmacología , Relación Dosis-Respuesta en la Radiación , Femenino , Genes p53 , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos/métodos , Radiación Ionizante , Tolueno/análogos & derivados , Tolueno/farmacología
11.
Cell Cycle ; 7(5): 656-64, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18256537

RESUMEN

During mouse fetal development, meiosis is initiated in female germ cells only, with male germ cells undergoing mitotic arrest. Retinoic acid (RA) is degraded by Cyp26b1 in the embryonic testis but not in the ovary where it initiates the mitosis/meiosis transition. However the role of RA status in fetal germ cell proliferation has not been elucidated. As expected, using organ cultures, we observed that addition of RA in 11.5 days post-conception (dpc) testes induced Stra8 expression and meiosis. Surprisingly, in 13.5 dpc testes although RA induced Stra8 expression it did not promote meiosis. On 11.5 and 13.5 dpc, RA prevented male germ cell mitotic arrest through PI3K signaling. Therefore 13.5 dpc testes appeared as an interesting model to investigate RA effects on germ cell proliferation/differentiation independently of RA effect on the meiosis induction. At this stage, RA delayed SSEA-1 extinction, p63gamma expression and DNA hypermethylation which normally occur in male mitotic arrested germ cells. In vivo, in the fetal male gonad, germ cells cease their proliferation and loose SSEA-1 earlier than in female gonad and RA administration maintained male germ cell proliferation. Lastly, inhibition of endogenous Cyp26 activity in 13.5 dpc cultured testes also prevented male germ cell mitotic arrest. Our data demonstrate that the reduction of RA levels, which occurs specifically in the male fetal gonad and was known to block meiosis initiation, is also necessary to allow the establishment of the germ cell mitotic arrest and the correct further differentiation of the fetal germ cells along the male pathway.


Asunto(s)
Feto/citología , Mitosis/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Tretinoina/farmacología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/enzimología , Femenino , Feto/efectos de los fármacos , Feto/enzimología , Masculino , Meiosis/efectos de los fármacos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Transducción de Señal/efectos de los fármacos , Espermatozoides/enzimología , Testículo/enzimología , Tretinoina/administración & dosificación
12.
Toxicol Sci ; 99(1): 234-43, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17569695

RESUMEN

There is now compelling evidence that inappropriate exposure to estrogen during fetal or neonatal life could affect adult reproductive functions because the testis is sensitive to estrogens during specific periods of its development. Therefore, we investigated the effects of exogenous estrogens on gametogenesis and steroidogenesis during fetal and neonatal testicular development in the rat. We used in vitro systems, organ cultures, and dispersed testicular cell cultures, which allow the development of fetal and neonatal germ cells (gonocytes) and Leydig cells. Exogenous estrogens inhibited testosterone production in dispersed testicular cell cultures throughout fetal life, but this inhibition was observed only in the early fetal stages in organ culture. By using an aromatase inhibitor (letrozole, Novartis Pharma AG), we showed that the inhibitory effect of exogenous estrogens on testosterone production is masked in the whole testis at later stages (20.5 days postconception) due essentially to local production of estrogens. In both systems, additions of high concentrations (10(-6) M) of 17beta-estradiol or diethylstilbestrol decreased the number of gonocytes during the first fetal proliferative period but not during the neonatal period. Letrozole was without effect, suggesting that the aging-related loss of responsiveness of gonocytes is not due to any aromatase activity in the gonocytes.


Asunto(s)
Dietilestilbestrol/toxicidad , Estradiol/toxicidad , Desarrollo Fetal/efectos de los fármacos , Organogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Animales Recién Nacidos , Inhibidores de la Aromatasa/farmacología , Recuento de Células , Células Cultivadas , Relación Dosis-Respuesta a Droga , Edad Gestacional , Letrozol , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/patología , Masculino , Nitrilos/farmacología , Técnicas de Cultivo de Órganos , Ratas , Ratas Wistar , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Testículo/patología , Testículo/fisiopatología , Triazoles/farmacología
13.
J Cell Physiol ; 210(1): 87-98, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16998800

RESUMEN

The fetal and neonatal development of male germ cells (gonocytes) is a poorly understood but crucial process for establishing fertility. In rodents, gonocytes go through two phases of proliferation accompanied by apoptosis and separated by a quiescent period during the end of fetal development. P63 is a member of the P53 gene family that yields six isoforms. We detected only the p63 protein and no p53 and p73 in the nucleus of the gonocytes of mouse testes. We report for the first time the ontogeny of each p63 mRNA isoform during testis development. We observed a strong expression of p63gamma mRNA and protein when gonocytes are in the quiescent period. In vitro treatment with retinoic acid prevented gonocytes from entering the quiescent period and was correlated with a reduced production of p63gamma isoform mRNA. We investigated the function of p63 by studying the testicular phenotype of P63-null mice. P63 invalidation slightly, but significantly increased the number of gonocytes counted during the quiescent period. As P63-null animals die at birth we used an original organ culture that mimicked neonatal in vivo development to study further the testicular development. P63 invalidation resulted in a sharply increased number of gonocytes during the culture period due to a decrease in spontaneous apoptosis with no change in proliferation. P63 invalidation also caused abnormal morphologies in the germ cells that were also found in P63(+/-) adult male mice. Thus, p63 appears as an important regulator of germ cell development.


Asunto(s)
Apoptosis , Regulación del Desarrollo de la Expresión Génica , Fosfoproteínas/metabolismo , Espermatogénesis , Espermatogonias/metabolismo , Testículo/metabolismo , Transactivadores/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Edad Gestacional , Inmunohistoquímica , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis/efectos de los fármacos , Proteínas Nucleares/metabolismo , Técnicas de Cultivo de Órganos , Fosfoproteínas/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatogénesis/efectos de los fármacos , Espermatogonias/citología , Espermatogonias/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Testículo/embriología , Transactivadores/genética , Tretinoina/farmacología , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
14.
Med Sci (Paris) ; 21(12): 1083-8, 2005 Dec.
Artículo en Francés | MEDLINE | ID: mdl-16324650

RESUMEN

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. 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.


Asunto(s)
Testículo/anomalías , Testículo/efectos de los fármacos , Xenobióticos/efectos adversos , Estrógenos/efectos adversos , Humanos , Recién Nacido , Masculino , Testículo/embriología , Testículo/crecimiento & desarrollo
15.
Endocrinology ; 146(5): 2454-61, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15661855

RESUMEN

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.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Estrógenos/fisiología , Feto/citología , Células Intersticiales del Testículo/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Dietilestilbestrol/farmacología , Receptor alfa de Estrógeno/deficiencia , Femenino , Células Intersticiales del Testículo/química , Hormona Luteinizante/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fosfoproteínas/genética , Embarazo , ARN Mensajero/análisis , Esteroide 17-alfa-Hidroxilasa/genética , Testículo/embriología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testosterona/metabolismo
16.
Endocrinology ; 145(7): 3395-403, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15044378

RESUMEN

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.


Asunto(s)
Estrógenos/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Testículo/citología , Testículo/embriología , Animales , Animales Recién Nacidos , Apoptosis , Peso Corporal , División Celular , Línea Celular , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Células Germinativas/citología , Células Germinativas/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ratones Noqueados , Embarazo , Células de Sertoli/citología , Células de Sertoli/metabolismo , Testículo/metabolismo , Testosterona/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...