Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753613

RESUMEN

Fetal growth plays a role in programming of adult cardiometabolic disorders, which in men, are associated with lowered testosterone levels. Fetal growth and fetal androgen exposure can also predetermine testosterone levels in men, although how is unknown, because the adult Leydig cells (ALCs) that produce testosterone do not differentiate until puberty. To explain this conundrum, we hypothesized that stem cells for ALCs must be present in the fetal testis and might be susceptible to programming by fetal androgen exposure during masculinization. To address this hypothesis, we used ALC ablation/regeneration to identify that, in rats, ALCs derive from stem/progenitor cells that express chicken ovalbumin upstream promoter transcription factor II. These stem cells are abundant in the fetal testis of humans and rodents, and lineage tracing in mice shows that they develop into ALCs. The stem cells also express androgen receptors (ARs). Reduction in fetal androgen action through AR KO in mice or dibutyl phthalate (DBP) -induced reduction in intratesticular testosterone in rats reduced ALC stem cell number by ∼40% at birth to adulthood and induced compensated ALC failure (low/normal testosterone and elevated luteinizing hormone). In DBP-exposed males, this failure was probably explained by reduced testicular steroidogenic acute regulatory protein expression, which is associated with increased histone methylation (H3K27me3) in the proximal promoter. Accordingly, ALCs and ALC stem cells immunoexpressed increased H3K27me3, a change that was also evident in ALC stem cells in fetal testes. These studies highlight how a key component of male reproductive development can fundamentally reprogram adult hormone production (through an epigenetic change), which might affect lifetime disease risk.


Asunto(s)
Células Madre Adultas/fisiología , Andrógenos/fisiología , Desarrollo Fetal/fisiología , Células Intersticiales del Testículo/fisiología , Células Madre Adultas/efectos de los fármacos , Animales , Callithrix , Linaje de la Célula/fisiología , Dibutil Ftalato/toxicidad , Femenino , Desarrollo Fetal/efectos de los fármacos , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/fisiología , Humanos , Técnicas In Vitro , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Embarazo , Ratas , Ratas Transgénicas , Ratas Wistar , Receptores Androgénicos/deficiencia , Receptores Androgénicos/genética , Receptores Androgénicos/fisiología , Regeneración , Testículo/embriología , Testículo/fisiología , Testosterona/deficiencia , Testosterona/fisiología
2.
Hum Reprod ; 28(4): 886-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23321215

RESUMEN

STUDY QUESTION: Is perinatal germ cell (GC) differentiation in the marmoset similar to that in the human? SUMMARY ANSWER: In a process comparable with the human, marmoset GC differentiate rapidly after birth, losing OCT4 expression after 5-7 weeks of age during mini-puberty. WHAT IS KNOWN ALREADY: Most of our understanding about perinatal GC development derives from rodents, in which all gonocytes (undifferentiated GC) co-ordinately lose expression of the pluripotency factor OCT4 and stop proliferating in late gestation. Then after birth these differentiated GC migrate to the basal lamina and resume proliferation prior to the onset of spermatogenesis. In humans, fetal GC differentiation occurs gradually and asynchronously and OCT4(+) GC persist into perinatal life. Failure to switch off OCT4 in GC perinatally can lead to development of carcinoma in situ (CIS), the precursor of testicular germ cell cancer (TGCC), for which there is no animal model. Marmosets show similarities to the human, but systematic evaluation of perinatal GC development in this species is lacking. Similarity, especially for loss of OCT4 expression, would support use of the marmoset as a model for the human and for studying CIS origins. STUDY DESIGN, SIZE AND DURATION: Testis tissues were obtained from marmosets (n = 4-10 per age) at 12-17 weeks' gestation and post-natal weeks 0.5, 2.5, 5-7, 14 and 22 weeks, humans at 15-18 weeks' gestation (n = 5) and 4-5 weeks of age (n = 4) and rats at embryonic day 21.5 (e21.5) (n = 3) and post-natal days 4, 6 and 8 (n = 4 each). PARTICIPANTS/MATERIALS, SETTING AND METHODS: Testis sections from fetal and post-natal marmosets, humans and rats were collected and immunostained for OCT4 and VASA to identify undifferentiated and differentiated GC, respectively, and for Ki67, to identify proliferating GC. Stereological quantification of GC numbers, differentiation (% OCT4(+) GC) and proliferation were performed in perinatal marmosets and humans. Quantification of GC position within seminiferous cords was performed in marmosets, humans and rats. MAIN RESULTS AND ROLE OF CHANCE: The total GC number increased 17-fold from birth to 22 post-natal weeks in marmosets; OCT4(+) and VASA(+) GC proliferated equally in late gestation and early post-natal life. The percentage of OCT4(+) GC fell from 54% in late fetal life to <0.5% at 2.5 weeks of age and none were detected after 5-7 weeks in marmosets. In humans, the percentage of OCT4(+) GC also declined markedly during the equivalent period. In marmosets, GC had begun migrating to the base of seminiferous cords at ∼22 weeks of age, after the loss of GC OCT4 expression. LIMITATIONS, REASONS FOR CAUTION: There is considerable individual variation between marmosets. Although GC development in marmosets and humans was similar, there are differences with respect to proliferation during fetal life. The number of human samples was limited. WIDER IMPLICATIONS OF THE FINDINGS: The similarities in testicular GC differentiation between marmosets and humans during the perinatal period, and their differences from rodents, suggest that the marmoset may be a useful model for studying the origins of CIS, with relevance for the study of TGCC. STUDY FUNDING/COMPETING INTERESTS: This work was supported by Grant G33253 from the Medical Research Council, UK. No external funding was sought and there are no competing interests.


Asunto(s)
Callithrix/fisiología , Diferenciación Celular , Espermatozoides/citología , Animales , Carcinoma in Situ/patología , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Ratas , Testículo/citología , Testículo/metabolismo , Testículo/patología
3.
Biol Reprod ; 81(6): 1083-92, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19587329

RESUMEN

Cluster analysis at Postnatal Day 8-20 of putative androgen-regulated genes in mice with Sertoli cell-selective knockout of the androgen receptor (SCARKO) has pinpointed three genes (Spinlw1, Gpd1, Drd4) with an expression pattern strongly resembling that of Rhox5, the definitive Sertoli cell (SC) androgen-regulated gene. We used organotypic testis cultures from Day 8 mice to study control of these genes by (anti)androgens and follicle-stimulating hormone (FSH). Testis morphology and androgen induction of the studied genes were preserved for 48 h. Preincubation with ketoconazole for 24 h to block endogenous androgen production, followed by 24-h incubation with the synthetic androgen R1881, resulted in 45-, 5-, 19-, and 6-fold induction of mRNA levels of Rhox5, Spinlw1, Gpd1, and Drd4, respectively. However, noticeable differences in control of the studied genes were observed. Rhox5 and Spinlw1 were fully induced by R1881 in the continuous (48 h) presence of ketoconazole, whereas only marginal effects were observed on expression of Gpd1 and Drd4. Similarly, FSH only marginally affected expression of Rhox5 and Spinlw1, whereas it markedly increased Gpd1 and Drd4 expression. Explant cultures of SCARKO testes confirmed the differential effects of FSH on the studied genes and, for Gpd1, showed that the effect did not depend on a functional androgen receptor in SC, whereas this was essential for the effects of FSH on Drd4. In conclusion, organotypic cultures represent the first in vitro approach to preserving androgen responsiveness of putative SC-expressed genes. This approach facilitates detailed analysis of their regulation in ways not possible in vivo.


Asunto(s)
Andrógenos/metabolismo , Técnicas de Cultivo de Órganos/métodos , Células de Sertoli/metabolismo , Testículo/metabolismo , Análisis de Varianza , Andrógenos/farmacología , Animales , Apoptosis/fisiología , Medios de Cultivo Condicionados , Perfilación de la Expresión Génica , Glicerolfosfato Deshidrogenasa/genética , Glicerolfosfato Deshidrogenasa/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos , Proteínas Inhibidoras de Proteinasas Secretoras , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Dopamina D4/genética , Receptores de Dopamina D4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/efectos de los fármacos , Testículo/citología , Testículo/efectos de los fármacos , Testosterona/metabolismo , Testosterona/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Hum Reprod ; 24(9): 2244-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19491204

RESUMEN

BACKGROUND: Fetal exposure of male rats to some phthalates induces reproductive abnormalities, raising concerns for similar effects in humans. In order to address this in a more appropriate animal model, the aim of the present studies was to investigate the effect of fetal/neonatal exposure to monobutyl phthalate (MBP) in a non-human primate, the marmoset. In particular, to determine if exposure resulted in effects at birth, or in adulthood, similar to those in male rats, and whether there was evidence for induction of carcinoma-in-situ (CIS) or testicular germ cell tumours (TGCT). METHODS: Pregnant female marmosets were dosed from approximately 7-15 weeks gestation with 500 mg/kg/day MBP and male offspring studied at birth (1-5 days; n = 6) or in adulthood (n = 5). In another study, newborn males (n = 5 co-twins) were dosed with 500 mg/kg/day MBP for 14 days, commencing at approximately 4 days of age. RESULTS: Fetal exposure of marmosets to MBP did not affect gross testicular morphology, reproductive tract development or testosterone levels at birth, nor were germ cell number and proliferation, Sertoli cell number or germ:Sertoli cell ratio affected. In two of six MBP-exposed animals, unusual clusters of undifferentiated germ cells were found, but their significance is unclear. Neonatal MBP treatment did not affect germ cell numbers or differentiation. Fetal exposure to MBP did not affect testis size/morphology, germ cell numbers or fertility in adulthood. There was no evidence for CIS or TGCT. CONCLUSIONS: Fetal exposure of marmosets to MBP does not measurably affect testis development/function or cause testicular dysgenesis, and no effects emerge by adulthood. Some effects on germ cell development were found, but these were inconsistent and of uncertain significance.


Asunto(s)
Exposición Materna , Ácidos Ftálicos/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Callithrix , Femenino , Células Germinativas/efectos de los fármacos , Masculino , Embarazo , Células de Sertoli/efectos de los fármacos
5.
Endocrinology ; 149(10): 5280-7, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18566125

RESUMEN

Fetal androgen action is an important determinant of Sertoli cell (SC) number at birth. Androgens "program" reproductive tract development in rats between embryonic d (e) 15.5 and e17.5 ("male programming window"), and this is reflected for life by anogenital distance (AGD). We investigated if androgen regulation of SC number/proliferation was also programmed by androgens in this window. Pregnant rats were treated in various fetal time windows with vehicle (control) or 500 mg/kg.d di(n-butyl) phthalate (DBP), which suppresses fetal intratesticular testosterone (ITT). ITT and SC number/proliferation index were determined at e17.5 or e21.5; AGD was also determined at e21.5. In controls, SC number increased 11-fold and ITT by 10-fold from e17.5-e21.5. In animals exposed daily to DBP from e13.5, SC number was reduced by approximately 50% at e21.5, but increased 6-fold, as did ITT, from e17.5-e21.5; DBP had no effect on ITT at e15.5, reduced ITT by 50% at e17.5, and by more than 75% at e19.5-21.5. DBP exposure just in the male programming window did not alter SC number at e17.5 or 21.5 but reduced AGD. DBP treatment beyond e19.5 caused major reductions in SC number/proliferation index and ITT at e21.5. Only DBP treatments that included the male programming window led to reduced AGD at e21.5, but SC number was clearly not programmed in this window. Nevertheless, testis weight correlated highly (P<0.001) with AGD at e21.5, and postnatal d 25 and 90 in animals exposed in utero to vehicle or DBP (e13.5-e21.5). Thus, AGD may predict adult testis size but probably not through a direct relationship with SC number.


Asunto(s)
Andrógenos/metabolismo , Células de Sertoli/citología , Testículo/citología , Testículo/embriología , Testosterona/metabolismo , Factores de Edad , Animales , Recuento de Células , División Celular/efectos de los fármacos , División Celular/fisiología , Dibutil Ftalato/farmacología , Femenino , Inmunohistoquímica , Masculino , Tamaño de los Órganos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Maduración Sexual/fisiología , Testículo/metabolismo
6.
Int J Androl ; 31(2): 103-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17877717

RESUMEN

A testicular dysgenesis-like syndrome is induced in rats by fetal exposure to di(n-butyl) phthalate (DBP). A key feature of this is the formation of focal dysgenetic areas comprising malformed seminiferous cords/tubules and intratubular Leydig cells (ITLC), but how and why these arise remains unclear. The present study has used combinations of cell-specific markers and immunohistochemistry to address this. The results show that focal dysgenetic areas and ITLC first appear postnatally at 4-10 days of age, but this only occurs in treatment groups in which formation of fetal Leydig cell aggregation is induced between e17.5 and e21.5. Extreme variability in the formation and size of the Leydig cell aggregates probably accounts for the equally extreme variation in occurrence and size of focal dysgenetic areas postnatally. DBP-induced fetal Leydig cell aggregation traps Sertoli and other cells within the aggregates, but it is unclear why this happens nor why cords fail to form prenatally in these cell mixtures but do elsewhere in the fetal testis. The present studies show that differentiation of the fetal Leydig cells is drastically delayed at e15.5 after DBP exposure, which may be indicative of a wider delay in testis cell development and organisation, and this might account for some of the unexplained findings.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Testiculares/patología , Testículo/crecimiento & desarrollo , Animales , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar
7.
Environ Health Perspect ; 126(4): 047006, 2018 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-29665328

RESUMEN

BACKGROUND: Analgesic exposure during pregnancy may affect aspects of fetal gonadal development that are targeted by endocrine disruptors. OBJECTIVES: We investigated whether therapeutically relevant doses of acetaminophen and ibuprofen affect germ cell (GC) development in human fetal testes/ovaries using in vitro and xenograft approaches. METHODS: First-trimester human fetal testes/ovaries were cultured and exposed to acetaminophen or ibuprofen (7 d). Second-trimester human fetal testes were xenografted into mice and exposed to acetaminophen (1 or 7 d), or ibuprofen (7 d). To determine mechanism of action, a human GC tumor­derived cell line (NTera2) exhibiting fetal GC characteristics was used in addition to in vitro and in vivo rat models. RESULTS AND DISCUSSION: Gonocyte (TFAP2C+) number was reduced relative to controls in first-trimester human fetal testes exposed in vitro to acetaminophen (-28%) or ibuprofen (-22%) and also in ovaries exposed to acetaminophen (-43%) or ibuprofen (-49%). Acetaminophen exposure reduced gonocyte number by 17% and 30% in xenografted second-trimester human fetal testes after treatment of host mice for 1 or 7 d, respectively. NTera2 cell number was reduced following exposure to either analgesic or prostaglandin E2 (PGE2) receptor antagonists, whereas PGE2 agonists prevented acetaminophen-induced reduction in NTera2 cell number. Expression of GC pluripotency genes, and genes that regulate DNA/histone methylation, also differed from controls following analgesic and PGE2 receptor antagonist exposures. Gene expression changes were observed in rat fetal testis/ovary cultures and after in vivo acetaminophen exposure of pregnant rats. For example, expression of the epigenetic regulator TET1, was increased following exposure to acetaminophen in human NTera2 cells, rat fetal testis/ovary cultures, and in fetal testes and ovaries after in vivo exposure of pregnant rats, indicating translatability across experimental models and species. CONCLUSIONS: Our results demonstrate evidence of PGE2-mediated effects of acetaminophen and ibuprofen on GC/NTera2 cells, which raises concerns about analgesic use during human pregnancy that warrant further investigation. https://doi.org/10.1289/EHP2307.


Asunto(s)
Acetaminofén/efectos adversos , Diferenciación Celular/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Células Germinativas/efectos de los fármacos , Ibuprofeno/efectos adversos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Xenoinjertos , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Desnudos , Ovario/efectos de los fármacos , Embarazo , Primer Trimestre del Embarazo , Segundo Trimestre del Embarazo , Testículo/efectos de los fármacos
8.
Environ Health Perspect ; 115(3): 390-6, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17431488

RESUMEN

BACKGROUND: Certain phthalates can impair Leydig cell distribution and steroidogenesis in the fetal rat in utero, but it is unknown whether similar effects might occur in the human. OBJECTIVES: Our aim in this study was to investigate the effects of di(n-butyl) phthalate (DBP), or its metabolite monobutyl phthalate (MBP), on testosterone production and Leydig cell aggregation (LCA) in fetal testis explants from the rat and human, and to compare the results with in vivo findings for DBP-exposed rats. We also wanted to determine if DBP/MBP affects testosterone production in vivo in the neonatal male marmoset. METHODS: Fetal testis explants obtained from the rat [gestation day (GD)19.5] and from the human (15-19 weeks of gestation) were cultured for 24-48 hr with or without human chorionic gonadotropin (hCG) or 22R-hydroxycholesterol (22R-OH), and with or without DBP/MBP. Pregnant rats and neonatal male marmosets were dosed with 500 mg/kg/day DBP or MBP. RESULTS: Exposure of rats in utero to DBP (500 mg/kg/day) for 48 hr before GD21.5 induced major suppression of intratesticular testosterone levels and cytochrome P450 side chain cleavage enzyme (P450scc) expression; this short-term treatment induced LCA, but was less marked than longer term (GD13.5-20.5) DBP treatment. In vitro, MBP (10(-3) M) did not affect basal or 22R-OH-stimulated testosterone production by fetal rat testis explants but slightly attenuated hCG-stimulated steroidogenesis; MBP induced minor LCA in vitro. None of these parameters were affected in human fetal testis explants cultured with 10(-3) M MBP for up to 48 hr. Because the in vivo effects of DBP/MBP were not reproduced in vitro in the rat, the absence of MBP effects in vitro on fetal human testes is inconclusive. In newborn (Day 2-7) marmosets, administration of a single dose of 500 mg/kg MBP significantly (p = 0.019) suppressed blood testosterone levels 5 hr later. Similar treatment of newborn co-twin male marmosets for 14 days resulted in increased Leydig cell volume per testis (p = 0.011), compared with co-twin controls; this is consistent with MBP-induced inhibition of steroidogenesis followed by compensatory Leydig cell hyperplasia/hypertrophy. CONCLUSIONS: These findings suggest that MBP/DBP suppresses steroidogenesis by fetal-type Leydig cells in primates as in rodents, but this cannot be studied in vitro.


Asunto(s)
Dibutil Ftalato/toxicidad , Ácidos Ftálicos/toxicidad , Testículo/efectos de los fármacos , Animales , Animales Recién Nacidos , Callithrix , Agregación Celular/efectos de los fármacos , Femenino , Humanos , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Exposición Materna , Embarazo , Ratas , Ratas Wistar , Testículo/embriología , Testículo/metabolismo , Testosterona/metabolismo
9.
JCI Insight ; 2(6): e91204, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28352662

RESUMEN

The testicular dysgenesis syndrome (TDS) hypothesis, which proposes that common reproductive disorders of newborn and adult human males may have a common fetal origin, is largely untested. We tested this hypothesis using a rat model involving gestational exposure to dibutyl phthalate (DBP), which suppresses testosterone production by the fetal testis. We evaluated if induction of TDS via testosterone suppression is restricted to the "masculinization programming window" (MPW), as indicated by reduction in anogenital distance (AGD). We show that DBP suppresses fetal testosterone equally during and after the MPW, but only DBP exposure in the MPW causes reduced AGD, focal testicular dysgenesis, and TDS disorders (cryptorchidism, hypospadias, reduced adult testis size, and compensated adult Leydig cell failure). Focal testicular dysgenesis, reduced size of adult male reproductive organs, and TDS disorders and their severity were all strongly associated with reduced AGD. We related our findings to human TDS cases by demonstrating similar focal dysgenetic changes in testes of men with preinvasive germ cell neoplasia (GCNIS) and in testes of DBP-MPW animals. If our results are translatable to humans, they suggest that identification of potential causes of human TDS disorders should focus on exposures during a human MPW equivalent, especially if negatively associated with offspring AGD.


Asunto(s)
Disgenesia Gonadal/inducido químicamente , Enfermedades Testiculares/inducido químicamente , Animales , Dibutil Ftalato/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Exposición Materna , Plastificantes/toxicidad , Ratas
10.
Endocrinology ; 147(11): 5352-62, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16916955

RESUMEN

This study investigated effects of in utero exposure [embryonic day (e)13.5-e21.5] to di(n-butyl) phthalate (DBP) on fetal gonocytes and postnatal germ cell (GC) development in rats and focused on changes (delayed development) relevant to the postulated origins of human carcinoma-in situ cells. DBP treatment resulted in both early (e15.5-e17.5) and late (e19.5-e21.5) effects on gonocytes. The former involved delayed entry of proliferating gonocytes into quiescence, as indicated by prolongation/overexpression of octamer-binding transcription factor 3/4 and retinoblastoma protein phosphorylated at Ser 807/811 and Ki67 plus a 2- to 4-fold increase in gonocyte apoptosis. The late effect of DBP was to induce a greater than 10-fold increase in occurrence of multinucleated gonocytes. GC numbers in DBP-exposed males were reduced (P < 0.01) by 37, 53, 79, and 80% at e21.5 and postnatal d (d) 4, 8, and 15, respectively, with recovery to normal in scrotal testes between postnatal d 25 and 90. The DBP-induced decrease in GC numbers at d 4-8 was associated with delayed exit from quiescence, as indicated by retinoblastoma protein expression and a 28% reduction (P < 0.001) in GC proliferation index at d 6, although the latter was increased by 84% at d 25. The postnatal GC changes were associated with the early, but not late, effects of DBP on gonocytes as short-term DBP treatment from e19.5 to e20.5, induced multinucleated gonocytes as effectively as did treatment from e13.5 to e20.5, but did not reduce GC numbers on d 4. In conclusion, fetal DBP exposure delays normal GC development in both fetal (as early as e15.5) and postnatal life with the possibility of consequences for fertility.


Asunto(s)
Dibutil Ftalato/toxicidad , Feto/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Inmunohistoquímica , Antígeno Ki-67/análisis , Masculino , Factor 3 de Transcripción de Unión a Octámeros/análisis , Ratas , Ratas Wistar , Proteína de Retinoblastoma/análisis , Recuento de Espermatozoides , Espermatozoides/efectos de los fármacos , Espermatozoides/patología , Testículo/patología
11.
Sci Rep ; 6: 19789, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26813099

RESUMEN

Analgesics which affect prostaglandin (PG) pathways are used by most pregnant women. As germ cells (GC) undergo developmental and epigenetic changes in fetal life and are PG targets, we investigated if exposure of pregnant rats to analgesics (indomethacin or acetaminophen) affected GC development and reproductive function in resulting offspring (F1) or in the F2 generation. Exposure to either analgesic reduced F1 fetal GC number in both sexes and altered the tempo of fetal GC development sex-dependently, with delayed meiotic entry in oogonia but accelerated GC differentiation in males. These effects persisted in adult F1 females as reduced ovarian and litter size, whereas F1 males recovered normal GC numbers and fertility by adulthood. F2 offspring deriving from an analgesic-exposed F1 parent also exhibited sex-specific changes. F2 males exhibited normal reproductive development whereas F2 females had smaller ovaries and reduced follicle numbers during puberty/adulthood; as similar changes were found for F2 offspring of analgesic-exposed F1 fathers or mothers, we interpret this as potentially indicating an analgesic-induced change to GC in F1. Assuming our results are translatable to humans, they raise concerns that analgesic use in pregnancy could potentially affect fertility of resulting daughters and grand-daughters.


Asunto(s)
Analgésicos/farmacología , Células Germinativas/citología , Células Germinativas/efectos de los fármacos , Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Reproducción/efectos de los fármacos , Animales , Diferenciación Celular , Femenino , Feto , Masculino , Fenotipo , Embarazo , Prostaglandinas/metabolismo , Ratas
12.
Endocrinology ; 146(2): 613-23, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15539561

RESUMEN

Fetal exposure of male rats to di (n-butyl) phthalate (DBP) induces testicular changes remarkably similar to testicular dysgenesis syndrome in humans; these include induction of focal areas of dysgenetic tubules in otherwise normal testes. In searching for the fetal origins of the latter, we used image analysis to show that exposure to 500 mg/kg DBP [embryonic day (E)13.5-20.5)] caused abnormal aggregation of Leydig cells centrally in the fetal testis. This aggregation was not due to increase in Leydig cell number, and Leydig cell size was significantly reduced in DBP-exposed animals, as were testosterone levels and immunoexpression of P450 side-chain cleavage enzyme. The Leydig cell aggregates did not exhibit evidence of focal proliferation at E17.5-19.5. Using confocal microscopy and Leydig (3beta-hydroxysteroid dehydrogenase) and Sertoli (anti-Mullerian hormone) cell-specific markers, we show that fetal Leydig cell aggregates in DBP-exposed animals trap isolated Sertoli cells within them at E21.5. These areas of intermingled cells are still apparent on postnatal d 4, after cessation of DBP treatment, when they may form misshapen seminiferous cords that trap (intratubular) Leydig cells within them. These centrally located dysgenetic tubules contain germ cells in early puberty, but by adulthood they are Sertoli cell only, implying that presence of intratubular Leydig cells interferes with spermatogenesis. It is concluded that DBP-induced fetal Leydig cell aggregation may be a key event in formation of focal dysgenetic areas in the testis, and identification of the mechanisms underlying these events may give new insights into the fetal origins of testicular dysgenesis syndrome disorders in the human.


Asunto(s)
Anomalías Inducidas por Medicamentos/patología , Antagonistas de Andrógenos/toxicidad , Dibutil Ftalato/toxicidad , Células Intersticiales del Testículo/patología , Túbulos Seminíferos/anomalías , Animales , Agregación Celular/efectos de los fármacos , Femenino , Hiperplasia , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ratas , Ratas Wistar , Túbulos Seminíferos/patología
13.
Endocrinology ; 146(10): 4536-44, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16037377

RESUMEN

Cryptorchidism is a common reproductive abnormality, possibly resulting from abnormal hormone production/action by the fetal testis. Insulin-like factor 3 (Insl3) is thought to be involved in gubernaculum development and transabdominal testicular descent, but its importance is unclear, due partly to lack of suitable Insl3 antibodies. We generated (by genetic immunization) and validated a novel antirat Insl3 antibody, which we used to characterize immunoexpression of Insl3 in rat Leydig cells (LCs) from fetal life until adulthood and its relationship to cryptorchidism. Immunoexpression was strong on embryonic day (E) 17.5 and E19.5 and from 35 d of age onward but weak from E21.5 until puberty. Because in utero exposure to di (n-butyl) phthalate (DBP) induces cryptorchidism and suppresses Insl3 gene expression, we investigated Insl3 protein expression in fetal and adult rats exposed to 500 mg/kg.d DBP from E13.5 to E21.5. Expression on E17.5 and E19.5 decreased dramatically after DBP exposure, but there was no consistent correlation between this suppression and abnormal testis position. We also compared expression of Insl3 and P450 side-chain cleavage enzyme in fetal testes from rats exposed in utero to DBP or flutamide (50 mg/kg.d). DBP treatment suppressed expression of both P450 side-chain cleavage enzyme and Insl3 at E19.5, but flutamide exposure had no effect on either protein, demonstrating that Insl3 expression in fetal rat LCs is not androgen regulated. In adult rats, Insl3 expression was suppressed in 80% of cryptorchid and 50% of scrotal testes from rats exposed to DBP, suggesting that prenatal DBP exposure also leads to maldevelopment/malfunction of the adult LC population in some animals.


Asunto(s)
Criptorquidismo/inducido químicamente , Dibutil Ftalato/toxicidad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Testículo/embriología , Animales , Criptorquidismo/embriología , Criptorquidismo/patología , Cartilla de ADN , Femenino , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/fisiología , Masculino , Microscopía Fluorescente , Embarazo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo
14.
Endocrinology ; 146(9): 4117-26, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15919750

RESUMEN

It is established that androgens and unidentified Sertoli cell (SC)-derived factors can influence the development of adult Leydig cells (LC) in rodents, but the mechanisms are unclear. We evaluated adult LC development and function in SC-selective androgen receptor (AR) knockout (SCARKO) and complete AR knockout (ARKO) mice. In controls, LC number increased 26-fold and LC size increased by approximately 2-fold between 12 and 140 d of age. LC number in SCARKOs was normal on d 12, but was reduced by more than 40% at later ages, although LC were larger and contained more lipid droplets and mitochondria than control LC by adulthood. ARKO LC number was reduced by up to 83% at all ages compared with controls, and LC size did not increase beyond d 12. Serum LH and testosterone levels and seminal vesicle weights were comparable in adult SCARKOs and controls, whereas LH levels were elevated 8-fold in ARKOs, although testosterone levels appeared normal. Immunohistochemistry and quantitative PCR for LC-specific markers indicated steroidogenic function per LC was probably increased in SCARKOs and reduced in ARKOs. In SCARKOs, insulin-like factor-3 and estrogen sulfotransferase (EST) mRNA expression were unchanged and increased 3-fold, respectively, compared with controls, whereas the expression of both was reduced more than 90% in ARKOs. Changes in EST expression, coupled with reduced platelet-derived growth factor-A expression, are potential causes of altered LC number and function in SCARKOs. These results show that loss of androgen action on SC has major consequences for LC development, and this could be mediated indirectly via platelet-derived growth factor-A and/or estrogens/EST.


Asunto(s)
Células Intersticiales del Testículo/fisiología , Células Intersticiales del Testículo/ultraestructura , Receptores Androgénicos/genética , Células de Sertoli/citología , Células de Sertoli/fisiología , Factores de Edad , Andrógenos/fisiología , Animales , Recuento de Células , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Testículo/citología , Testículo/fisiología
15.
J Endocrinol ; 184(1): 107-17, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15642788

RESUMEN

Neonatal treatment of male rats with diethylstilboestrol (DES) or a gonadotrophin-releasing hormone antagonist (GnRHa) reduces final Sertoli cell number, an effect presumed to occur via suppression of follicle-stimulating hormone (FSH). As both treatments also suppress androgen action, we investigated whether androgens and oestrogens might affect Sertoli cell nuclear volume/number in the rat using single or combined treatments that differentially affected FSH, testosterone and oestrogen (DES) levels. Neonatal treatment with flutamide (50 mg/ kg) significantly reduced Sertoli cell nuclear volume/ number per testis and blood inhibin-B levels at day 18, despite elevating FSH levels; this treatment also exacerbated the reduction in Sertoli cell nuclear volume per testis induced by treatment with 0.1 mug DES without affecting FSH levels. Treatment with 0.1 mug DES on its own also reduced Sertoli cell nuclear volume per testis without affecting FSH/testosterone levels, but co-administration of 0.1 mug DES+GnRHa, which suppressed FSH and testosterone levels, resulted in a markedly greater effect. Treatment with GnRHa alone or 10 mug DES alone grossly suppressed FSH and testosterone levels and reduced Sertoli cell nuclear volume/number per testis by approximately 60%, but co-administration of the two treatments had no greater effect than either alone. Co-administration of testosterone esters with 10 mug DES partially prevented the 10 mug DES-induced reduction in Sertoli cell nuclear volume per testis, and normalized FSH levels. In all treatment groups, plasma levels of inhibin-B paralleled changes in Sertoli cell nuclear volume/number per testis, but treatments that suppressed FSH levels (GnRHa, 10 mug DES) caused a proportionately greater reduction (approximately 90%) in inhibin-B levels than in Sertoli cell nuclear volume/number (50-60%). Germ cell volume per unit Sertoli cell was reduced in animals treated with 10 mug DES alone or in those treated with 0.1 mug DES plus either flutmaide or GnRHa, but otherwise remained relatively constant between treatment groups. It is concluded that, in the neonatal rat, (1) endogenous androgens, as well as FSH, play a physiological role in increasing Sertoli cell number, (2) exogenous oestrogen exposure can decrease Sertoli cell number without altering FSH levels, (3) these effects probably share a common pathway and (4) blood inhibin-B provides a robust indicator of change in Sertoli cell number.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Estrógenos no Esteroides/farmacología , Flutamida/farmacología , Hormona Liberadora de Gonadotropina/farmacología , Células de Sertoli/efectos de los fármacos , Animales , Animales Recién Nacidos , Biomarcadores/sangre , División Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Dietilestilbestrol/farmacología , Hormona Folículo Estimulante/antagonistas & inhibidores , Hormona Folículo Estimulante/sangre , Inhibinas/sangre , Masculino , Ratas , Ratas Wistar , Testosterona/sangre
16.
Environ Health Perspect ; 113(11): 1580-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16263515

RESUMEN

The purpose of this study was to evaluate whether experimental exposure of pregnant sheep to a mixture of environmental chemicals added to pasture as sewage sludge (n = 9 treated animals) exerted effects on fetal testis development or function; application of sewage sludge was undertaken so as to maximize exposure of the ewes to its contents. Control ewes (n = 9) were reared on pasture treated with an equivalent amount of inorganic nitrogenous fertilizer. Treatment had no effect on body weight of ewes, but it reduced body weight by 12-15% in male (n = 12) and female (n = 8) fetuses on gestation day 110. In treated male fetuses (n = 11), testis weight was significantly reduced (32%), as were the numbers of Sertoli cells (34% reduction), Leydig cells (37% reduction), and gonocytes (44% reduction), compared with control fetuses (n = 8). Fetal blood levels of testosterone and inhibin A were also reduced (36% and 38%, respectively) in treated compared with control fetuses, whereas blood levels of luteinizing hormone and follicle-stimulating hormone were unchanged. Based on immunoexpression of anti-Müllerian hormone, cytochrome P450 side chain cleavage enzyme, and Leydig cell cytoplasmic volume, we conclude that the hormone changes in treated male fetuses probably result from the reduction in somatic cell numbers. This reduction could result from fetal growth restriction in male fetuses and/or from the lowered testosterone action; reduced immunoexpression of alpha-smooth muscle actin in peritubular cells and of androgen receptor in testes of treated animals supports the latter possibility. These findings indicate that exposure of the developing male sheep fetus to real-world mixtures of environmental chemicals can result in major attenuation of testicular development and hormonal function, which may have consequences in adulthood.


Asunto(s)
Contaminantes Ambientales/efectos adversos , Fertilizantes/efectos adversos , Aguas del Alcantarillado/efectos adversos , Testículo/efectos de los fármacos , Animales , Femenino , Desarrollo Fetal/efectos de los fármacos , Hormona Folículo Estimulante/sangre , Inhibinas/sangre , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/sangre , Masculino , Exposición Materna , Embarazo , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Ovinos/embriología , Ovinos/metabolismo , Testículo/embriología , Testículo/metabolismo , Testosterona/sangre
17.
Endocrinology ; 156(1): 24-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25375036

RESUMEN

Androgen action during the fetal masculinization programming window (MPW) determines the maximum potential for growth of androgen-dependent organs (eg, seminal vesicles, prostate, penis, and perineum) and is reflected in anogenital distance (AGD). As such, determining AGD in postnatal life has potential as a lifelong easily accessible biomarker of overall androgen action during the MPW. However, whether the perineum remains androgen responsive in adulthood and thus responds plastically to perturbed androgen drive remains unexplored. To determine this, we treated adult male rats with either the antiandrogen flutamide or the estrogen diethylstilbestrol (DES) for 5 weeks, followed by a 4-week washout period of no treatment. We determined AGD and its correlate anogenital index (AGI) (AGD relative to body weight) at weekly intervals across this period and compared these with normal adult rats (male and female), castrated male rats, and appropriate vehicle controls. These data showed that, in addition to reducing circulating testosterone and seminal vesicle weight, castration significantly reduced AGD (by ∼17%), demonstrating that there is a degree of plasticity in AGD in adulthood. Flutamide treatment increased circulating testosterone yet also reduced seminal vesicle weight due to local antagonism of androgen receptor. Despite this suppression, surprisingly, flutamide treatment had no effect on AGD at any time point. In contrast, although DES treatment suppressed circulating testosterone and reduced seminal vesicle weight, it also induced a significant reduction in AGD (by ∼11%), which returned to normal 1 week after cessation of DES treatment. We conclude that AGD in adult rats exhibits a degree of plasticity, which may be mediated by modulation of local androgen/estrogen action. The implications of these findings regarding the use of AGD as a lifelong clinical biomarker of fetal androgen action are discussed.


Asunto(s)
Andrógenos/fisiología , Perineo/crecimiento & desarrollo , Maduración Sexual/fisiología , Antagonistas de Andrógenos/farmacología , Animales , Biomarcadores , Estudios de Cohortes , Estrógenos/fisiología , Femenino , Flutamida/farmacología , Hormona Luteinizante/sangre , Masculino , Orquiectomía , Perineo/embriología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Distribución Aleatoria , Ratas , Ratas Wistar , Testosterona/sangre
18.
Environ Health Perspect ; 123(3): 223-30, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514601

RESUMEN

BACKGROUND: Phthalate exposure induces germ cell effects in the fetal rat testis. Although experimental models have shown that the human fetal testis is insensitive to the steroidogenic effects of phthalates, the effects on germ cells have been less explored. OBJECTIVES: We sought to identify the effects of phthalate exposure on human fetal germ cells in a dynamic model and to establish whether the rat is an appropriate model for investigating such effects. METHODS: We used immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction to examine Sertoli and germ cell markers on rat testes and human fetal testis xenografts after exposure to vehicle or di(n-butyl) phthalate (DBP). Our study included analysis of germ cell differentiation markers, proliferation markers, and cell adhesion proteins. RESULTS: In both rat and human fetal testes, DBP exposure induced similar germ cell effects, namely, germ cell loss (predominantly undifferentiated), induction of multinucleated gonocytes (MNGs), and aggregation of differentiated germ cells, although the latter occurred rarely in the human testes. The mechanism for germ cell aggregation and MNG induction appears to be loss of Sertoli cell-germ cell membrane adhesion, probably due to Sertoli cell microfilament redistribution. CONCLUSIONS: Our findings provide the first comparison of DBP effects on germ cell number, differentiation, and aggregation in human testis xenografts and in vivo in rats. We observed comparable effects on germ cells in both species, but the effects in the human were muted compared with those in the rat. Nevertheless, phthalate effects on germ cells have potential implications for the next generation, which merits further study. Our results indicate that the rat is a human-relevant model in which to explore the mechanisms for germ cell effects.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dibutil Ftalato/toxicidad , Células Germinativas/efectos de los fármacos , Sustancias Peligrosas/toxicidad , Testículo/efectos de los fármacos , Animales , Feto/efectos de los fármacos , Humanos , Inmunohistoquímica , Masculino , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Testículo/embriología , Trasplante Heterólogo
19.
Sci Transl Med ; 7(288): 288ra80, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25995226

RESUMEN

Most common male reproductive disorders are linked to lower testosterone exposure in fetal life, although the factors responsible for suppressing fetal testosterone remain largely unknown. Protracted use of acetaminophen during pregnancy is associated with increased risk of cryptorchidism in sons, but effects on fetal testosterone production have not been demonstrated. We used a validated xenograft model to expose human fetal testes to clinically relevant doses and regimens of acetaminophen. Exposure to a therapeutic dose of acetaminophen for 7 days significantly reduced plasma testosterone (45% reduction; P = 0.025) and seminal vesicle weight (a biomarker of androgen exposure; 18% reduction; P = 0.005) in castrate host mice bearing human fetal testis xenografts, whereas acetaminophen exposure for just 1 day did not alter either parameter. Plasma acetaminophen concentrations (at 1 hour after the final dose) in exposed host mice were substantially below those reported in humans after a therapeutic oral dose. Subsequent in utero exposure studies in rats indicated that the acetaminophen-induced reduction in testosterone likely results from reduced expression of key steroidogenic enzymes (Cyp11a1, Cyp17a1). Our results suggest that protracted use of acetaminophen (1 week) may suppress fetal testosterone production, which could have adverse consequences. Further studies are required to establish the dose-response and treatment-duration relationships to delineate the maximum dose and treatment period without this adverse effect.


Asunto(s)
Acetaminofén/toxicidad , Testículo/efectos de los fármacos , Testosterona/biosíntesis , Animales , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Femenino , Supervivencia de Injerto/efectos de los fármacos , Xenoinjertos , Humanos , Masculino , Ratones , Orquiectomía , Tamaño de los Órganos , Embarazo , Ratas , Medición de Riesgo , Vesículas Seminales/efectos de los fármacos , Vesículas Seminales/crecimiento & desarrollo , Vesículas Seminales/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo , Testículo/embriología , Testículo/metabolismo , Testosterona/sangre , Factores de Tiempo
20.
Endocrinology ; 143(12): 4797-808, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12446607

RESUMEN

This study tested the hypothesis that testis/reproductive tract abnormalities induced in the rat by neonatal treatment with diethylstilbestrol (DES) result from disturbance of the androgen-estrogen balance. Male rats were treated neonatally with a dose of DES (0.1 micro g) that induced either no or small effects on its own or with a dose (10 micro g) that induced major reproductive tract abnormalities. To allow quantification, the abnormalities chosen for study were distension of the rete testis and efferent ducts and reduction in epithelial cell height in the efferent ducts and vas deferens. To alter the androgen-estrogen balance, other rats were treated with DES (0.1 micro g) in combination with a treatment to suppress either androgen production [GnRH antagonist (GnRHa)] or androgen action (flutamide); other rats were treated with GnRHa or flutamide alone. Testosterone levels were measured to verify the effects of treatment. Combined administration of DES (0.1 micro g) plus GnRHa or flutamide induced significantly greater distension/overgrowth of the rete testis and efferent ducts (ED) and a reduction in epithelial cell height of the ED than did DES (0.1 micro g) administered alone. Neither GnRHa nor flutamide affected rete or ED distension when administered alone, but both significantly reduced ED epithelial cell height. Neonatal treatment with bisphenol-A (100 micro g) with or without GnRHa had no significant effect on any of these parameters. In contrast to the ED, a reduction in cell height of the vas deferens was induced to an equal extent by DES (10 micro g), DES (0.1 micro g) with GnRHa, and GnRHa alone, suggesting greater sensitivity of this tissue to both androgen and estrogen action. The induction of major abnormalities in rats treated with DES (10 micro g) was coincident with loss of androgen receptor immunoexpression in affected tissues. Reduced androgen receptor immunoexpression was also induced by combined treatment with DES (0.1 micro g) plus GnRHa or flutamide, whereas treatment with any of these compounds alone had no or only minor effects. These findings suggest that reduced androgen action sensitizes the reproductive tract to estrogens, demonstrating that the balance in action between androgens and estrogens, rather than their absolute levels, may be of fundamental importance in determining normal or abnormal development of some regions of the male reproductive tract.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Andrógenos/fisiología , Animales Recién Nacidos/crecimiento & desarrollo , Dietilestilbestrol/administración & dosificación , Estrógenos/fisiología , Reproducción/fisiología , Animales , Tamaño de la Célula , Células Epiteliales/citología , Receptor alfa de Estrógeno , Flutamida/farmacología , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Masculino , Ratas , Ratas Wistar , Receptores Androgénicos/análisis , Receptores de Estrógenos/análisis , Testículo/citología , Testículo/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testosterona/sangre , Conducto Deferente/citología , Conducto Deferente/efectos de los fármacos , Conducto Deferente/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA