Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Environ Health Perspect ; 126(4): 047006, 2018 04 16.
Article in English | MEDLINE | ID: mdl-29665328

ABSTRACT

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.


Subject(s)
Acetaminophen/adverse effects , Cell Differentiation/drug effects , Fetal Development/drug effects , Germ Cells/drug effects , Ibuprofen/adverse effects , Animals , Dose-Response Relationship, Drug , Female , Heterografts , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Ovary/drug effects , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Testis/drug effects
2.
JCI Insight ; 2(6): e91204, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28352662

ABSTRACT

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.


Subject(s)
Gonadal Dysgenesis/chemically induced , Testicular Diseases/chemically induced , Animals , Dibutyl Phthalate/toxicity , Disease Models, Animal , Female , Humans , Male , Maternal Exposure , Plasticizers/toxicity , Rats
3.
Sci Rep ; 6: 19789, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26813099

ABSTRACT

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.


Subject(s)
Analgesics/pharmacology , Germ Cells/cytology , Germ Cells/drug effects , Maternal Exposure , Prenatal Exposure Delayed Effects , Reproduction/drug effects , Animals , Cell Differentiation , Female , Fetus , Male , Phenotype , Pregnancy , Prostaglandins/metabolism , Rats
4.
Sci Transl Med ; 7(288): 288ra80, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995226

ABSTRACT

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.


Subject(s)
Acetaminophen/toxicity , Testis/drug effects , Testosterone/biosynthesis , Animals , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Female , Graft Survival/drug effects , Heterografts , Humans , Male , Mice , Orchiectomy , Organ Size , Pregnancy , Rats , Risk Assessment , Seminal Vesicles/drug effects , Seminal Vesicles/growth & development , Seminal Vesicles/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Testis/embryology , Testis/metabolism , Testosterone/blood , Time Factors
5.
Endocrinology ; 156(1): 24-31, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25375036

ABSTRACT

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.


Subject(s)
Androgens/physiology , Perineum/growth & development , Sexual Maturation/physiology , Androgen Antagonists/pharmacology , Animals , Biomarkers , Cohort Studies , Estrogens/physiology , Female , Flutamide/pharmacology , Luteinizing Hormone/blood , Male , Orchiectomy , Perineum/embryology , Pregnancy , Prenatal Exposure Delayed Effects , Random Allocation , Rats , Rats, Wistar , Testosterone/blood
6.
Environ Health Perspect ; 123(3): 223-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25514601

ABSTRACT

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.


Subject(s)
Cell Differentiation/drug effects , Dibutyl Phthalate/toxicity , Germ Cells/drug effects , Hazardous Substances/toxicity , Testis/drug effects , Animals , Fetus/drug effects , Humans , Immunohistochemistry , Male , Rats , Real-Time Polymerase Chain Reaction , Testis/embryology , Transplantation, Heterologous
7.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24753613

ABSTRACT

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.


Subject(s)
Adult Stem Cells/physiology , Androgens/physiology , Fetal Development/physiology , Leydig Cells/physiology , Adult Stem Cells/drug effects , Animals , Callithrix , Cell Lineage/physiology , Dibutyl Phthalate/toxicity , Female , Fetal Development/drug effects , Fetal Stem Cells/drug effects , Fetal Stem Cells/physiology , Humans , In Vitro Techniques , Leydig Cells/drug effects , Luteinizing Hormone/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Pregnancy , Rats , Rats, Transgenic , Rats, Wistar , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Receptors, Androgen/physiology , Regeneration , Testis/embryology , Testis/physiology , Testosterone/deficiency , Testosterone/physiology
8.
PLoS One ; 8(5): e62556, 2013.
Article in English | MEDLINE | ID: mdl-23671609

ABSTRACT

Recent studies have established that masculinization of the male reproductive tract is programmed by androgens in a critical fetal 'masculinization programming window' (MPW). What is peculiar to androgen action during this period is, however, unknown. Studies from 20 years ago in mice implicated prostaglandin (PG)-mediation of androgen-induced masculinization, but this has never been followed up. We therefore investigated if PGs might mediate androgen effects in the MPW by exposing pregnant rats to indomethacin (which blocks PG production by inhibiting cyclooxygenase activity) during this period and then examining if androgen production or action (masculinization) was affected. Pregnant rats were treated with indomethacin (0.8 mg/kg/day; e15.5-e18.5) to encompass the MPW. Indomethacin exposure decreased fetal bodyweight (e21.5), testis weight (e21.5) and testicular PGE2 (e17.5, e21.5), but had no effect on intratesticular testosterone (ITT; e17.5) or anogenital index (AGI; e21.5). Postnatally, AGI, testis weight and blood testosterone were unaffected by indomethacin exposure and no cryptorchidism or hypospadias occurred. Penis length was normal in indomethacin-exposed animals at Pnd25 but was reduced by 26% (p<0.001) in adulthood, an effect that is unexplained. Our results demonstrate that indomethacin can effectively decrease intra-testicular PGE2 level. However, the resulting male phenotype does not support a role for PGs in mediating androgen-induced masculinization during the MPW in rats. The contrast with previous mouse studies is unexplained but may reflect a species difference.


Subject(s)
Cryptorchidism/chemically induced , Cyclooxygenase Inhibitors/toxicity , Hypospadias/chemically induced , Indomethacin/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Cryptorchidism/metabolism , Dinoprostone/metabolism , Female , Fetal Development/drug effects , Humans , Hypospadias/metabolism , Male , Organ Size/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar , Testis/drug effects , Testis/metabolism , Testis/pathology , Testosterone/metabolism
9.
PLoS One ; 8(4): e61726, 2013.
Article in English | MEDLINE | ID: mdl-23620786

ABSTRACT

In rodents, in utero exposure to exogenous estrogens including diethylstilboestrol (DES) results in major suppression of steroidogenesis in fetal testes. Whether similar effects occur in the human fetal testis is equivocal. Based on the results of the rodent studies, we hypothesised that exposure of human fetal testes to DES would result in a reduction in testosterone production. We show, using a xenograft approach, that testosterone production is not reduced in human fetal testis following DES exposure. Human fetal testes (15-19 weeks' gestation, n = 6) were xenografted into castrate male nude mice which were then treated for 35 days with vehicle or 100 µg/kg DES three times a week. For comparison, similar treatment was applied to pregnant rats from e13.5-e20.5 and effects on fetal testes evaluated at e21.5. Xenograft testosterone production was assessed by measuring host seminal vesicle (SV) weights as an indirect measure over the entire grafting period, and single measurement of serum testosterone at termination. Human fetal testis xenografts showed similar survival in DES and vehicle-exposed hosts. SV weight (44.3 v 26.6 mg, p = 0.01) was significantly increased in DES compared to vehicle-exposed hosts, respectively, indicating an overall increase in xenograft testosterone production over the grafting period, whilst serum testosterone at termination was unchanged. In contrast intra-testicular testosterone levels were reduced by 89%, in fetal rats exposed to DES. In rats, DES effects are mediated via Estrogen Receptor α (ESR1). We determined ESR1 protein and mRNA expression in human and rat fetal testis. ESR1 was expressed in rat, but not in human, fetal Leydig cells. We conclude that human fetal testis exposure to DES does not impair testosterone production as it does in rats, probably because ESR1 is not expressed in human fetal Leydig cells. This indicates that DES exposure is likely to pose minimal risk to masculinization of the human fetus.


Subject(s)
Diethylstilbestrol/pharmacology , Fetus/metabolism , Testis/embryology , Testis/transplantation , Testosterone/biosynthesis , Transplantation, Heterologous , Animals , Diethylstilbestrol/administration & dosage , Embryo, Mammalian/drug effects , Endometrium/drug effects , Endometrium/metabolism , Estrogen Receptor alpha/metabolism , Female , Fetus/drug effects , Graft Survival/drug effects , Humans , Male , Mice , Mice, Nude , Pregnancy , Rats , Rats, Wistar , Testis/drug effects , Testis/metabolism
10.
Hum Reprod ; 28(4): 886-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23321215

ABSTRACT

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.


Subject(s)
Callithrix/physiology , Cell Differentiation , Spermatozoa/cytology , Animals , Carcinoma in Situ/pathology , Cell Proliferation , Disease Models, Animal , Humans , Male , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , Rats , Testis/cytology , Testis/metabolism , Testis/pathology
11.
PLoS One ; 7(5): e37064, 2012.
Article in English | MEDLINE | ID: mdl-22615892

ABSTRACT

Reproductive disorders that are common/increasing in prevalence in human males may arise because of deficient androgen production/action during a fetal 'masculinization programming window'. We identify a potentially important role for Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII) in Leydig cell (LC) steroidogenesis that may partly explain this. In rats, fetal LC size and intratesticular testosterone (ITT) increased ~3-fold between e15.5-e21.5 which associated with a progressive decrease in the percentage of LC expressing COUP-TFII. Exposure of fetuses to dibutyl phthalate (DBP), which induces masculinization disorders, dose-dependently prevented the age-related decrease in LC COUP-TFII expression and the normal increases in LC size and ITT. We show that nuclear COUP-TFII expression in fetal rat LC relates inversely to LC expression of steroidogenic factor-1 (SF-1)-dependent genes (StAR, Cyp11a1, Cyp17a1) with overlapping binding sites for SF-1 and COUP-TFII in their promoter regions, but does not affect an SF-1 dependent LC gene (3ß-HSD) without overlapping sites. We also show that once COUP-TFII expression in LC has switched off, it is re-induced by DBP exposure, coincident with suppression of ITT. Furthermore, other treatments that reduce fetal ITT in rats (dexamethasone, diethylstilbestrol (DES)) also maintain/induce LC nuclear expression of COUP-TFII. In contrast to rats, in mice DBP neither causes persistence of fetal LC COUP-TFII nor reduces ITT, whereas DES-exposure of mice maintains COUP-TFII expression in fetal LC and decreases ITT, as in rats. These findings suggest that lifting of repression by COUP-TFII may be an important mechanism that promotes increased testosterone production by fetal LC to drive masculinization. As we also show an age-related decline in expression of COUP-TFII in human fetal LC, this mechanism may also be functional in humans, and its susceptibility to disruption by environmental chemicals, stress and pregnancy hormones could explain the origin of some human male reproductive disorders.


Subject(s)
COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Leydig Cells/metabolism , Leydig Cells/physiology , Male Urogenital Diseases/physiopathology , Animals , Binding Sites/drug effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dibutyl Phthalate/adverse effects , Female , Fetus/drug effects , Fetus/metabolism , Fetus/physiology , Leydig Cells/drug effects , Male , Male Urogenital Diseases/genetics , Male Urogenital Diseases/metabolism , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , RNA, Messenger/genetics , Rats , Rats, Wistar , Rodentia , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Testosterone/metabolism
12.
Endocrinology ; 151(6): 2868-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20392824

ABSTRACT

Androgens may be important regulators of Sertoli cell (SC) proliferation perinatally, with implications for the testicular dysgenesis syndrome (TDS) hypothesis. Fetal exposure of rats to 500 mg/kg . d di(n-butyl) phthalate (DBP) reduces fetal testosterone production and SC number at birth, but SC number recovers to normal by postnatal d (Pnd)25. It is unclear when and how SC proliferation is affected prenatally by DBP exposure or when and how postnatal compensation occurs. This study addressed these questions and investigated whether continued maternal exposure to DBP or to flutamide from Pnd1-Pnd15 could prevent SC number compensation, because this would have implications for how sperm counts might be lowered in TDS. DBP exposure attenuated SC proliferation by 7-18% throughout embryonic d (e)15.5-e21.5 (P < 0.05 at e21.5). After birth, SC proliferation increased significantly (>1.5-fold) between Pnd6 and Pnd10 in prenatally DBP-exposed animals, explaining the compensation. Continued maternal administration of DBP after birth attenuated (19% reduction) SC number compensation at Pnd25 and maternal administration of flutamide (100 mg/kg . d) to prenatally DBP-exposed animals was even more effective (42% reduction), suggesting the postnatal compensatory increase in SC proliferation after prenatal DBP exposure is androgen dependent. SC maturation (Pnd25) was unaffected, based on analysis of expression of key proteins, but lumen formation/expansion was attenuated in parallel with treatment-induced reduction in SC number. Our results provide further evidence that perinatal SC proliferation is androgen dependent and, importantly, show that similar exposure of mothers to antiandrogenic chemicals before birth and during lactation reduces final SC number, with implications for the origin of low sperm counts in TDS.


Subject(s)
Dibutyl Phthalate/pharmacology , Flutamide/pharmacology , Sertoli Cells/cytology , Sertoli Cells/drug effects , Animals , Female , Fetus/drug effects , Fetus/physiology , Immunohistochemistry , Male , Organ Size/drug effects , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , SOX9 Transcription Factor/metabolism , Sertoli Cells/metabolism , Testis/cytology , Testis/drug effects , Testis/metabolism , Testis/physiology
13.
Biol Reprod ; 81(6): 1083-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19587329

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Organ Culture Techniques/methods , Sertoli Cells/metabolism , Testis/metabolism , Analysis of Variance , Androgens/pharmacology , Animals , Apoptosis/physiology , Culture Media, Conditioned , Gene Expression Profiling , Glycerolphosphate Dehydrogenase/genetics , Glycerolphosphate Dehydrogenase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Male , Mice , Organ Size/drug effects , Proteinase Inhibitory Proteins, Secretory , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Receptors, Dopamine D4/genetics , Receptors, Dopamine D4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Testis/cytology , Testis/drug effects , Testosterone/metabolism , Testosterone/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Hum Reprod ; 24(9): 2244-54, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19491204

ABSTRACT

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.


Subject(s)
Maternal Exposure , Phthalic Acids/toxicity , Prenatal Exposure Delayed Effects/pathology , Testis/drug effects , Testis/growth & development , Animals , Animals, Newborn , Callithrix , Female , Germ Cells/drug effects , Male , Pregnancy , Sertoli Cells/drug effects
15.
Endocrinology ; 149(10): 5280-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18566125

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Sertoli Cells/cytology , Testis/cytology , Testis/embryology , Testosterone/metabolism , Age Factors , Animals , Cell Count , Cell Division/drug effects , Cell Division/physiology , Dibutyl Phthalate/pharmacology , Female , Immunohistochemistry , Male , Organ Size , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sexual Maturation/physiology , Testis/metabolism
16.
Biol Reprod ; 78(2): 352-60, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17928633

ABSTRACT

Pregnancy exposure to di(n-butyl) phthalate (DBP) in rats induces a testicular dysgenesislike syndrome (TDS) in male offspring. Earlier studies suggested altered Sertoli cell development/maturation may result, especially in testes that become cryptorchid. This study quantitatively assessed Sertoli cell numerical and functional development in DBP-exposed rats and compared (unilaterally) cryptorchid and scrotal testes. Pregnant rats were gavaged with 500 mg/kg/day DBP or corn oil from embryonic (E) Days 13.5 to 21.5. Male offspring were sampled on E21.5 or Postnatal Day 6, 10, 15, 25, or 90. Sertoli cell number in DBP-exposed males was reduced by approximately 50% at E21.5 but recovered to normal by Days 25-90, accompanied by significant changes in plasma inhibin B and testosterone levels. Sertoli cell maturational development in DBP-exposed males, assessed using five protein markers (anti-müllerian hormone, cytokeratin, androgen receptor, CDKN1B, and Nestin), was largely normal, with some evidence of delayed maturation. However, in adulthood, Sertoli cells (SC) in areas lacking germ cells (Sertoli cell-only [SCO] tubules) often exhibited immature features, especially in cryptorchid testes. Sertoli cells in DBP-exposed animals supported fewer germ cells during puberty, but this normalized in scrotal testes by adulthood. Scrotal and especially cryptorchid testes from DBP-exposed animals exhibited abnormalities (SCO tubules, focal dysgenetic areas) at all postnatal ages. Cryptorchid testes from DBP-exposed animals exhibited more Sertoli cell abnormalities at Day 25 compared with scrotal testes, perhaps indicating more severe underlying Sertoli cell malfunction in these testes. Our findings support the concept of altered Sertoli cell development in TDS, especially in cryptorchid testes, but show that maturational defects in Sertoli cells in adulthood most commonly reflect secondary dedifferentiation in absence of germ cells.


Subject(s)
Cryptorchidism/pathology , Gonadal Dysgenesis/pathology , Sertoli Cells/pathology , Testis/abnormalities , Testis/pathology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Count , Cryptorchidism/chemically induced , Dibutyl Phthalate/toxicity , Disease Models, Animal , Follicle Stimulating Hormone/blood , Gonadal Dysgenesis/chemically induced , Inhibins/blood , Male , Organ Size , Plasticizers/toxicity , Proteins/analysis , Proteins/metabolism , Rats , Rats, Wistar , Sertoli Cells/drug effects , Sertoli Cells/physiology , Spermatogonia/cytology , Spermatogonia/physiology , Syndrome , Testosterone/blood
17.
Int J Androl ; 31(2): 103-11, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17877717

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Testicular Diseases/pathology , Testis/growth & development , Animals , Immunohistochemistry , Male , Rats , Rats, Wistar
18.
Environ Health Perspect ; 115(3): 390-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17431488

ABSTRACT

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.


Subject(s)
Dibutyl Phthalate/toxicity , Phthalic Acids/toxicity , Testis/drug effects , Animals , Animals, Newborn , Callithrix , Cell Aggregation/drug effects , Female , Humans , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Maternal Exposure , Pregnancy , Rats , Rats, Wistar , Testis/embryology , Testis/metabolism , Testosterone/metabolism
19.
Endocrinology ; 147(11): 5352-62, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16916955

ABSTRACT

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.


Subject(s)
Dibutyl Phthalate/toxicity , Fetus/drug effects , Spermatogenesis/drug effects , Testis/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Female , Immunohistochemistry , Ki-67 Antigen/analysis , Male , Octamer Transcription Factor-3/analysis , Rats , Rats, Wistar , Retinoblastoma Protein/analysis , Sperm Count , Spermatozoa/drug effects , Spermatozoa/pathology , Testis/pathology
20.
Int J Androl ; 29(1): 148-54; discussion 181-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16466534

ABSTRACT

Foetal exposure of male rats to di(n-butyl) phthalate (DBP) induces testicular changes similar to testicular dysgenesis syndrome in humans, including the formation of focal 'dysgenetic areas' within post-natal testes, surrounded by otherwise normal tubules exhibiting complete spermatogenesis. We hypothesize that these dysgenetic areas form when Sertoli (and other) cells are 'trapped' during the abnormal formation of large Leydig cell (LC) clusters in foetal life and by post-natal day (d) 4 these groups of intermingled cells attempt to form seminiferous tubules. It is likely that the malformed tubules resulting correspond to the dysgenetic areas evident in later life. This also provides a plausible explanation for the occurrence of LCs within seminiferous cords/tubules in or bordering the dysgenetic areas. In our previous studies intratubular LCs (ITLCs) were identified by immunostaining for 3beta-hydroxysteroid dehydrogenase (3beta-HSD), the definitive LC cytoplasmic marker. However, the possibility remained that the 'presumptive' ITLCs were in fact Sertoli cells that had aberrantly gained the ability to express 3beta-HSD. Therefore, the aim of the present study was to fully characterize the ITLCs induced by in utero DBP exposure in d25 rats using a number of LC- (3beta-HSD, P450 side-chain cleavage enzyme, insulin-like factor 3, oestrogen receptor alpha) and Sertoli cell- (vimentin, Wilm's tumour-1) specific markers. Our results show that ITLCs express all four LC-specific markers but do not express either of the Sertoli cell markers. It is therefore concluded that the ITLCs are bona fide LCs that are abnormally located within the seminiferous tubules of DBP-exposed rats in post-natal life.


Subject(s)
Androgen Antagonists/toxicity , Dibutyl Phthalate/toxicity , Disease Models, Animal , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects , Testis/abnormalities , Androgen Antagonists/administration & dosage , Animals , Dibutyl Phthalate/administration & dosage , Female , Humans , Leydig Cells/pathology , Male , Pregnancy , Rats , Rats, Wistar , Sertoli Cells/pathology , Testis/drug effects , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...