Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Arch Toxicol ; 97(2): 313-327, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36336711

RESUMO

Male reproduction is one of the primary health endpoints identified in rodent studies for some phthalates, such as DEHP (Bis(2-ethylhexyl) phthalate), DBP (Dibutyl phthalate), and BBP (Benzyl butyl phthalate). The reduction in testosterone level was used as an intermediate key event for grouping some phthalates and to establish a reference point for risk assessment. Phthalates, and specifically DEHP, are one of the chemicals for which the greatest number of non-monotonic dose responses (NMDRs) are observed. These NMDRs cover different endpoints and situations, often including testosterone levels. The presence of NMDR has been the subject of some debate within the area of chemical risk assessment, which is traditionally anchored around driving health-based guidance values for apical endpoints that typically follow a clear monotonic dose-response. The consequence of NMDR for chemical risk assessment has recently received considerable attention amongst regulatory agencies, which confirmed its relevance particularly for receptor-mediated effects. The present review explores the relationship between DEHP exposure and testosterone levels, investigating the biological plausibility of the observed NMDRs. The Adverse Outcome Pathway (AOP) concept is applied to integrate NMDRs into Key Event Relationships (KERs) for exploring a mechanistic understanding of initial key events and possibly associated reproductive and non-reproductive adverse outcomes.


Assuntos
Rotas de Resultados Adversos , Dietilexilftalato , Ácidos Ftálicos , Masculino , Animais , Dietilexilftalato/toxicidade , Ácidos Ftálicos/toxicidade , Dibutilftalato , Testosterona/metabolismo
2.
Int J Androl ; 35(3): 330-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22248293

RESUMO

Masculinization is programmed by androgen exposure during a masculinization programming window (MPW). Deficiency in MPW androgen action results in reduced size of all reproductive organs and anogenital distance (AGD) and reproductive disorders. Although timing of MPW closing has been defined, what determines 'opening' and 'closing' of the MPW remains unknown. To test whether initiation of testosterone production/action defines the opening of the window, we first demonstrated that androgen receptor mRNA and protein are expressed prior to the MPW, and then investigated whether masculinization could be advanced or enhanced by treating pregnant rats with either 1 or 10 mg/kg/day dihydrotestosterone (DHT) prior to (early window, EW; e11.5-e14.5) or during the MPW (e15.5-e18.5), and then evaluating offspring in foetal life (e18.5, e21.5), early puberty (day 25) or adulthood (∼day 75). DHT treatment did not affect pregnancy duration, birth, litter or pup size. DHT exposure in either time window did not advance foetal male development (Wolffian duct coiling) and had no effect on AGD, testis, penis and ventral prostate (VP) size at any age when measured; there was a tendency towards smaller penis size. In contrast, exposure of females to 10 mg DHT in either time window induced varying degrees of masculinization, including stabilization of the Wolffian duct and increased AGD (e21.5, Pnd25), VP formation, more male-like phallus structure, absence of nipples and vaginal opening and, in some adult females, gross fluid distension of the uterus (hydrometrocolpos); these effects were generally more pronounced after exposure in the MPW than in the EW. In conclusion, exposure of the male rat foetus to additional androgens prior to or during the MPW does not advance or enhance any measured parameter of reproductive development. Therefore, androgen availability plays no role in determining timing of the MPW. Susceptibility of the female reproductive system to androgens may precede the MPW.


Assuntos
Di-Hidrotestosterona/farmacologia , Genitália Masculina/embriologia , Processos de Determinação Sexual/fisiologia , Animais , Feminino , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/embriologia , Genitália Masculina/efeitos dos fármacos , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Receptores Androgênicos/biossíntese
3.
Int J Androl ; 35(5): 741-51, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22420564

RESUMO

Nitric oxide (NO) plays crucial roles in several physiological and pathological conditions. The iNOS isoform produces high levels of NO independent of intracellular calcium and, in the testis, which is expressed in Sertoli (SC), Leydig (LC) and germ cells. The testicular roles of NO are unclear, but it can inhibit LC testosterone production. Our aim was to evaluate the effects of iNOS deficiency on testis development in mice from late fetal life through early puberty. Therefore, testes from wild type (C57BCL/6) and iNOS(-/-) mice (B6.129P2- Nos2(tm1Lau) /J) were sampled at various ages between e18.5 and Pnd20 and evaluated by histological and stereological analyses; proliferating cells were labelled with (3)H-thymidine. At all ages, testis weight and anogenital index, a measure of fetal androgen exposure, were greater in iNOS-deficient mice than in wild type mice. At all ages after birth, iNOS-deficient mice exhibited increased (p < 0.05) SC number per testis, and this was accounted for by a higher SC proliferation index (p < 0.05) in iNOS-deficient mice, especially on Pnd1 and Pnd5. Similarly, LC number per testis was higher (p < 0.05) in iNOS(-/-) mice than in wild type at all post-natal ages. Highly positive and significant correlations were observed between the proliferation index for SC, LC and peritubular myoid cells on e18.5 and post-natally. Although lumen formation was slightly advanced in iNOS(-/-) mice, no obvious other effects on pubertal testis development were observed. These results imply that NO may normally constrain testis somatic cell development, especially SC, perhaps by limiting testosterone production. Removal of this constraint results in normal, but larger, testes with greater sperm production. Our data pinpoint the window of iNOS (NO) action on SC proliferation and raise the possibility that experimental manipulation of NO in early post-natal life could be used to enhance SC proliferation if this was deficient for any reason.


Assuntos
Óxido Nítrico Sintase Tipo II/deficiência , Células de Sertoli/citologia , Testículo/embriologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Intersticiais do Testículo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células de Sertoli/efeitos dos fármacos , Espermatogênese/fisiologia , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento
4.
Int J Androl ; 35(1): 25-40, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21651570

RESUMO

Testosterone synthesis depends on normal Leydig cell (LC) development, but the mechanisms controlling this development remain unclear. We recently demonstrated that androgen receptor (AR) ablation from a proportion of testicular peritubular myoid cells (PTM-ARKO) did not affect LC number, but resulted in compensated LC failure. The current study extends these investigations, demonstrating that PTM AR signalling is important for normal development, ultrastructure and function of adult LCs. Notably, mRNAs for LC markers [e.g. steroidogenic factor 1 (Nr5a1), insulin-like growth factor (Igf-1) and insulin-like factor 3 (Insl3)] were significantly reduced in adult PTM-ARKOs, but not all LCs were similarly affected. Two LC sub-populations were identified, one apparently 'normal' sub-population that expressed adult LC markers and steroidogenic enzymes as in controls, and another 'abnormal' sub-population that had arrested development and only weakly expressed INSL3, luteinizing hormone receptor, and several steroidogenic enzymes. Furthermore, unlike 'normal' LCs in PTM-ARKOs, the 'abnormal' LCs did not involute as expected in response to exogenous testosterone. Differential function of these LC sub-populations is likely to mean that the 'normal' LCs work harder to compensate for the 'abnormal' LCs to maintain normal serum testosterone. These findings reveal new paracrine mechanisms underlying adult LC development, which can be further investigated using PTM-ARKOs.


Assuntos
Diferenciação Celular , Células Intersticiais do Testículo/citologia , Receptores Androgênicos/metabolismo , Transdução de Sinais , Animais , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos
5.
Int J Androl ; 35(3): 317-29, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22150464

RESUMO

Exposure to ubiquitous, environmental chemicals (ECs) has been hypothesized as a cause for declining male reproductive health. Understanding the long-term effects of EC exposure on reproductive health in humans requires animal models and exposure to 'real life', environmentally relevant, mixtures during development, a life stage of particular sensitivity to ECs. The aim of this study was to evaluate the effects of in utero and post-natal exposure to environmentally relevant levels of ECs, via sewage sludge application to pasture, on the adult male sheep testis. Hormones, liver concentrations of candidate ECs and Sertoli and germ cell numbers in testes of adult rams that were exposed to ECs in sewage sludge in utero, and until weaning via maternal exposure, and post-weaning via grazing pastures fertilized with sewage sludge, were quantified. Evaluated as a single group, exposure to sludge ECs was without significant effect on most parameters. However, a more detailed study revealed that 5 of 12 sludge-exposed rams exhibited major spermatogenic abnormalities. These consisted of major reductions in germ cell numbers per testis or per Sertoli cell and more Sertoli cell-only tubules, when compared with controls, which did not show any such changes. The sludge-related spermatogenic changes in the five affected animals were significantly different from controls (p < 0.001); Sertoli cell number was unaffected. Hormone profiles and liver candidate EC concentrations were not measurably affected by exposure. We conclude that developmental exposure of male sheep to real-world mixtures of ECs can result in major reduction in germ cell numbers, indicative of impaired sperm production, in a proportion of exposed males. The individual-specific effects are presumed to reflect EC effects on a heterogeneous population in which some individuals may be more susceptible to adverse EC effects. Such effects of EC exposure in humans could have adverse consequences for sperm counts and fertility in some exposed males.


Assuntos
Esgotos/efeitos adversos , Espermatogênese/efeitos dos fármacos , Animais , Feminino , Humanos , Masculino , Saúde Reprodutiva , Síndrome de Células de Sertoli/epidemiologia , Carneiro Doméstico , Testículo/efeitos dos fármacos , Testículo/patologia
6.
Int J Androl ; 34(5 Pt 2): e386-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21332505

RESUMO

Environmental factors are implicated in increased incidence of human testicular germ-cell cancer (TGCC). TGCC has foetal origins and may be one component of a testicular dysgenesis syndrome (TDS). Certain phthalates induce TDS in rats, including effects on foetal germ cells (GC). As humans are widely exposed to phthalates, study of the effects of phthalates on foetal rat GC could provide an insight into the vulnerability of foetal GC to disruption by environmental factors, and thus to origins of TGCC. This study has therefore characterized foetal GC development in rats after in utero exposure to di(n-butyl) phthalate (DBP) with emphasis on GC numbers/proliferation, differentiation and time course for inducing effects. Pregnant rats were treated orally from embryonic day 13.5 (e13.5) with 500 mg/kg/day DBP for varying periods. GC number, proliferation, apoptosis, differentiation (loss of OCT4, DMRT1 expression, DMRT1 re-expression, GC migration) and aggregation were evaluated at various foetal and postnatal ages. DBP exposure reduced foetal GC number by ∼60% by e15.5 and prolonged GC proliferation, OCT4 and DMRT1 immunoexpression; these effects were induced in the period immediately after testis differentiation (e13.5-e15.5). In contrast, DBP-induced GC aggregation stemmed from late gestation effects (beyond e19.5). Foetal DBP exposure delayed postnatal resumption of GC proliferation, leading to bigger deficits in numbers, and delayed re-expression of DMRT1 and radial GC migration. Therefore, DBP differentially affects foetal GC in rats according to stage of gestation, effects that may be relevant to the human because of their nature (OCT4, DMRT1 effects) or because similar effects are demonstrable in vitro on human foetal testes (GC number). Identification of the mechanisms underlying these effects could give a new insight into environment-sensitive mechanisms in early foetal GC development that could potentially be relevant to TGCC origins.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dibutilftalato/toxicidade , Células Germinativas/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Feminino , Idade Gestacional , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Wistar
7.
Int J Androl ; 34(6 Pt 2): e578-86, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21631528

RESUMO

Experimental animal studies show that measurement of anogenital distance (AGD) and/or penis length may provide lifelong 'read-outs' of foetal androgen exposure during the masculinization programming window (MPW). However, variation in postnatal androgen exposure may complicate interpretation of such measurements. This is important to clarify if such measurements are to be applied to humans. The present aim was to evaluate effects of prenatal and/or postnatal manipulation of androgen production/action on growth of AGD and the penis in rats. Pregnant rats were treated daily before (e13.5-e21.5) and after birth (postnatal days 1-15) with either vehicle, 500 mg/kg di(n-butyl) phthalate (DBP) or 100 mg/kg flutamide (postnatal only) in prenatal + postnatal treatment combinations (N = 6 treatment combinations); DBP impairs androgen production whereas flutamide impairs androgen action. Male offspring were killed on postnatal day 8 (prepuberty), 25 (early puberty) or 90 (adulthood) when AGD was measured, the penis dissected out and its weight and length measured; plasma testosterone and ventral prostate weight were measured at day 90 to assess endogenous androgen exposure. In controls, penis length, girth and AGD increased 2.2-, 5.3-and 5.9-fold respectively from day 8 to day 90. Significant inhibition of penis growth and final length and girth was induced by treatments that inhibited postnatal androgen action. Conversely, growth and ultimate (adult) AGD was inhibited by prenatal inhibition of androgen production whereas postnatal androgen inhibition had negligible effect. Nevertheless, AGD and penis length were highly correlated at every age (R(2) > 0.33; p < 0.0001). However, altered endogenous androgen exposure may confound interpretation of changes in adults exposed prenatally/postnatally to DBP/flutamide. We conclude that AGD provides a lifelong guide to prenatal androgen exposure (in the MPW) whereas penis size reflects both prenatal + postnatal androgen exposure. At the group treatment level, prepubertal measurement of either AGD or penis size accurately predicts their size in adulthood.


Assuntos
Canal Anal/crescimento & desenvolvimento , Androgênios/fisiologia , Pênis/crescimento & desenvolvimento , Maturidade Sexual , Animais , Feminino , Masculino , Gravidez , Ratos , Ratos Wistar
8.
Int J Androl ; 34(6 Pt 2): e621-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21831234

RESUMO

Nitric oxide (NO) is produced via oxidation of l-arginine by nitric oxide synthases (NOSs), and is known as inducible (iNOS), neuronal, endothelial or testis-specific. Suggesting important functions for NOS in the normal rat and mouse testis, iNOS is reported to be constitutively expressed in Leydig cells (LC), Sertoli cells (SC) and germ cells. In our study, we sought to provide further insights into the roles of iNOS in the adult mouse testis using iNOS(-/-) mice. Perfusion-fixed testes from wild type (WT) and iNOS(-/-) mice were used for histological and stereological evaluations. Some of the mice had been injected with (3) H-thymidine to label proliferating cells and to determine the duration of spermatogenesis that was unaffected in iNOS(-/-) mice. Both LC nuclear volume and individual cell size were significantly decreased in iNOS(-/-) mice, but the total number of LC per testis was increased (p < 0.05) by approximately 16%. The number of SC per testis was strikingly increased (approximately twofold) in iNOS(-/-) mice, and testis weight and DSP per gram of testis (spermatogenic efficiency) were similarly increased. The anogenital distance was also significantly increased in iNOS(-/-) mice, and this key endpoint suggests that the augmentation observed for the SC number may be related to increased foetal T-exposure during the masculinization programming window. Compared with WT testes, the numbers of spermatocytes and spermatids and SC per tubule cross sections were significantly increased in iNOS(-/-) mice. Except for stages V-VI and VII-VIII, iNOS(-/-) mice exhibited approximately 3.5-fold fewer apoptotic germ cells than in WT mice. Taken together, our results provide new evidence that iNOS plays an important role in numerical and functional regulation of key somatic cells in the testis, which in turn impacts on germ cells and their survival and thus on daily sperm production.


Assuntos
Óxido Nítrico Sintase Tipo II/metabolismo , Células de Sertoli/citologia , Espermatogênese , Animais , Masculino , Camundongos , Camundongos Mutantes , Óxido Nítrico Sintase Tipo II/genética
9.
Int J Androl ; 33(2): 279-87, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20002220

RESUMO

We have shown previously that deficient androgen action within a masculinization programming window (MPW; e15.5-e18.5 in rats) is important in the origin of male reproductive disorders and in programming male reproductive organ size, but that androgen action postnatally may be important to achieve this size. To further investigate importance of the MPW, we used two rat models, in which foetal androgen production or action was impaired during the MPW by exposing in utero to either di(n-butyl) phthalate (DBP) or to flutamide. Reduced anogenital distance (AGD) was used as a monitor of androgen production/action during the MPW. Offspring were evaluated in early puberty (Pnd25) to establish if reproductive organ size was altered. The testes, penis, ventral prostate (VP) and seminal vesicles (SV) were weighed and penis length measured. Both DBP and flutamide exposure in the MPW significantly reduced penis, VP and SV size along with AGD at Pnd25; AGD and organ size were highly correlated. In DBP-, but not flutamide-, exposed animals, testis weight was also reduced and correlated with AGD. Intratesticular testosterone was also measured in control and DBP-exposed males during (e17.5) or after (e21.5) the MPW and related to AGD at e21.5. To evaluate the importance of postnatal androgen action in reproductive organ growth, the effect of combinations of prenatal and postnatal maternal treatments on AGD and penis size at Pnd25 was evaluated. In prenatally DBP-exposed animals, further postnatal exposure to either DBP or flutamide significantly reduced AGD and penis size in comparison with prenatal DBP exposure alone. In comparison, rats exposed postnatally to testosterone propionate after prenatal vehicle-exposure showed considerable increase in these parameters vs. controls. In conclusion, we show that the size of all male reproductive organs is programmed by androgen exposure in the MPW, but that growth towards this size is dependent on androgen action postnatally.


Assuntos
Androgênios/fisiologia , Dibutilftalato/toxicidade , Genitália Masculina/crescimento & desenvolvimento , Antagonistas de Androgênios/farmacologia , Androgênios/farmacologia , Animais , Animais Recém-Nascidos , Feminino , Flutamida/farmacologia , Genitália Masculina/efeitos dos fármacos , Disgenesia Gonadal/etiologia , Masculino , Tamanho do Órgão/efeitos dos fármacos , Pênis/efeitos dos fármacos , Pênis/crescimento & desenvolvimento , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Próstata/efeitos dos fármacos , Próstata/crescimento & desenvolvimento , Ratos , Ratos Wistar , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/crescimento & desenvolvimento , Diferenciação Sexual , Doenças Testiculares/etiologia , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento , Testículo/patologia , Testosterona/metabolismo , Propionato de Testosterona/farmacologia
10.
J Environ Monit ; 12(8): 1582-93, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20676422

RESUMO

Liver concentrations of selected pollutant classes were determined in groups of sheep fetuses and their dams, at 55 (Experiment 1) and 110 (Experiment 2) days of gestation (term = 145 d) following exposure, throughout their breeding lives and after mating, to pasture treated with either inorganic fertiliser (control, CC) or with sewage sludge (treated, TT). In a unique study designed to separate the respective contributions of environmental sources and mobilised tissue to the available EDC burden, in additional groups of animals, pollutant burdens at 110 days gestation were assessed following exposure to the respective treatments, either throughout their breeding lives until mating, but not thereafter (TC), or only between mating and slaughter (CT) (Experiment 3). With very few exceptions, maternal and fetal liver concentrations of diethylhexyl phthalate (DEHP) and selected polychlorinated biphenyls (PCBs), and polybrominated diphenyl ethers (PBDE) and polycyclic aromatic hydrocarbons (PAHs) were not significantly affected by sludge exposure in any group. In some cases, maternal and fetal tissue EDC concentrations were different but the differences were not consistent, and maternal and fetal concentrations of none of the classes of chemical were significantly correlated. It was not possible to identify a single chemical, or class of chemical, that may be responsible for previously observed physiological effects of exposure to sludge-treated pastures. It is concluded that exposure of sheep to pastures fertilised with sewage sludge was not associated with increased liver concentrations of EDCs, irrespective of the stage of development at which they were measured and of maternal tissue mobilisation and EDC release during gestation. Thus, retrospective measurements of EDC tissue burdens could not be used to accurately assess earlier fetal EDC insults.


Assuntos
Disruptores Endócrinos/metabolismo , Feto/metabolismo , Exposição Materna , Esgotos , Poluentes do Solo/metabolismo , Agricultura , Animais , Disruptores Endócrinos/análise , Feminino , Éteres Difenil Halogenados/análise , Éteres Difenil Halogenados/metabolismo , Bifenilos Policlorados/análise , Bifenilos Policlorados/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/análise , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Poluentes do Solo/análise , Eliminação de Resíduos Líquidos
11.
Endocr Dev ; 15: 101-134, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19293606

RESUMO

Infertility in the male is a potential complication of childhood cancer treatment for long-term survivors. The risk is dependent primarily on the treatment used, but also on the underlying disease. Chemotherapy (especially alkylating agents) and radiotherapy, even in low doses, may damage the seminiferous epithelium and impair spermatogenesis in both children and adults. Leydig cell function and testosterone production are generally preserved after chemotherapy and low dose radiotherapy, whilst larger doses of radiotherapy may result in hypogonadism. Patients treated with potentially gonadotoxic treatments require regular multidisciplinary follow-up including assessment of puberty and gonadal function. Currently the only option available for fertility preservation in young males treated for cancer is semen cryopreservation. For pre-pubertal patients, techniques for fertility preservation remain theoretical and as yet unproven. These include hormonal manipulation of the gonadal environment before treatment, germ cell transplantation and testis xenografting, which have all shown promise in a variety of animal studies. Refinement of these techniques requires investigations in relevant animal models. In the present chapter we include data which suggest that the common marmoset (Callithrix jacchus) monkey, a New World primate, exhibits important parallels with human testicular development and may help us to understand why the pre-pubertal testis is vulnerable to effects of cytotoxic therapy on future fertility.


Assuntos
Fertilidade , Infertilidade Masculina/prevenção & controle , Neoplasias/terapia , Preservação Biológica/métodos , Adulto , Animais , Criança , Irradiação Craniana/efeitos adversos , Citotoxinas/efeitos adversos , Citotoxinas/uso terapêutico , Fertilidade/efeitos dos fármacos , Fertilidade/fisiologia , Fertilidade/efeitos da radiação , Gônadas/efeitos dos fármacos , Gônadas/embriologia , Gônadas/crescimento & desenvolvimento , Gônadas/efeitos da radiação , Humanos , Infertilidade Masculina/etiologia , Masculino , Modelos Biológicos , Neoplasias/complicações , Reprodução/efeitos dos fármacos , Reprodução/efeitos da radiação , Maturidade Sexual/efeitos dos fármacos , Maturidade Sexual/fisiologia , Maturidade Sexual/efeitos da radiação
12.
Hum Reprod ; 23(12): 2755-65, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18694875

RESUMO

BACKGROUND: Testicular germ cell tumours (TGCT) are thought to originate from fetal germ cells that fail to differentiate normally, but no animal model for these events has been described. We evaluated the marmoset (Callithrix jacchus) as a model by comparing perinatal germ cell differentiation with that in humans. METHODS: Immunohistochemical profiling was used to investigate germ cell differentiation (OCT4, NANOG, AP-2gamma, MAGE-A4, VASA, NANOS-1) and proliferation (Ki67) in fetal and neonatal marmoset testes in comparison with the human and, to a lesser extent, the rat. RESULTS: In marmosets and humans, differentiation of gonocytes into spermatogonia is associated with the gradual loss of pluripotency markers such as OCT4 and NANOG, and the expression of germ cell-specific proteins such as VASA. This differentiation occurs asynchronously within individual cords during fetal and early postnatal life. This contrasts with rapid and synchronous germ cell differentiation within and between cords in the rat. Similarly, germ cell proliferation in the marmoset and human occurs throughout perinatal life, in contrast to rats in which proliferation ceases during this period. CONCLUSIONS: The marmoset provides a good model for normal human germ cell differentiation and proliferation. The perinatal marmoset may be a useful model in which to establish factors that lead to failure of normal germ cell differentiation and the origins of TGCT.


Assuntos
Callithrix/embriologia , Diferenciação Celular , Células Germinativas/citologia , Animais , Animais Recém-Nascidos , Proliferação de Células , RNA Helicases DEAD-box/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Masculino , Modelos Animais , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/biossíntese , Proteínas de Ligação a RNA/biossíntese , Ratos , Espermatogônias/metabolismo , Testículo/citologia , Testículo/embriologia , Fator de Transcrição AP-2/biossíntese
13.
Obes Rev ; 18(6): 673-686, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28371083

RESUMO

Animal models of maternal high fat diet (HFD) demonstrate perturbed offspring metabolism although the effects differ markedly between models. We assessed studies investigating metabolic parameters in the offspring of HFD fed mothers to identify factors explaining these inter-study differences. A total of 171 papers were identified, which provided data from 6047 offspring. Data were extracted regarding body weight, adiposity, glucose homeostasis and lipidaemia. Information regarding the macronutrient content of diet, species, time point of exposure and gestational weight gain were collected and utilized in meta-regression models to explore predictive factors. Publication bias was assessed using Egger's regression test. Maternal HFD exposure did not affect offspring birthweight but increased weaning weight, final bodyweight, adiposity, triglyceridaemia, cholesterolaemia and insulinaemia in both female and male offspring. Hyperglycaemia was found in female offspring only. Meta-regression analysis identified lactational HFD exposure as a key moderator. The fat content of the diet did not correlate with any outcomes. There was evidence of significant publication bias for all outcomes except birthweight. Maternal HFD exposure was associated with perturbed metabolism in offspring but between studies was not accounted for by dietary constituents, species, strain or maternal gestational weight gain. Specific weaknesses in experimental design predispose many of the results to bias.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hiperglicemia/metabolismo , Modelos Animais , Obesidade/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Aumento de Peso/fisiologia , Animais , Animais Recém-Nascidos/metabolismo , Feminino , Lactação/fisiologia , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Análise de Regressão , Desmame
15.
Sci Rep ; 6: 31857, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27550193

RESUMO

Experimental and epidemiological evidence demonstrate that ancestral diet might contribute towards offspring health. This suggests that nutrition may be able to modify genetic or epigenetic information carried by germ cells (GCs). To examine if a parental high fat diet (HFD) influences metabolic health in two generations of offspring, GC-eGFP Sprague Dawley rats were weaned onto HFD (45% fat) or Control Diet (CD; 10% fat). At 19 weeks, founders (F0) were bred with controls, establishing the F1 generation. HFD resulted in 9.7% and 14.7% increased weight gain in male and female F0 respectively. F1 offspring of HFD mothers and F1 daughters of HFD-fed fathers had increased weight gain compared to controls. F1 rats were bred with controls at 19 weeks to generate F2 offspring. F2 male offspring derived from HFD-fed maternal grandfathers exhibited increased adiposity, plasma leptin and luteinising hormone to testosterone ratio. Despite transmission via the founding male germline, we did not find significant changes in the F0 intra-testicular GC transcriptome. Thus, HFD consumption by maternal grandfathers results in a disrupted metabolic and reproductive hormone phenotype in grandsons in the absence of detectable changes in the intra-testicular GC transcriptome.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Obesidade/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Aumento de Peso , Adiposidade/genética , Animais , Feminino , Perfilação da Expressão Gênica , Leptina/sangue , Hormônio Luteinizante/sangue , Masculino , Obesidade/etiologia , Obesidade/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Ratos Sprague-Dawley , Fatores Sexuais , Testículo/metabolismo , Testosterona/sangue , Desmame
16.
J Neuroendocrinol ; 28(12)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27870155

RESUMO

We have shown that continuous maternal exposure to the complex mixture of environmental chemicals (ECs) found in human biosolids (sewage sludge), disrupts mRNA expression of genes crucial for development and long-term regulation of hypothalamic-pituitary gonadal (HPG) function in sheep. The present study investigated whether exposure to ECs only during preconceptional period or only during pregnancy perturbed key regulatory genes within the hypothalamus and pituitary gland and whether these effects were different from chronic (life-long) exposure to biosolid ECs. The findings demonstrate that the timing and duration of maternal EC exposure influences the subsequent effects on the foetal neuroendocrine system in a sex-specific manner. Maternal exposure prior to conception, or during pregnancy only, altered the expression of key foetal neuroendocrine regulatory systems such as gonadotrophin-releasing hormone and kisspeptin to a greater extent than when maternal exposure was 'life-long'. Furthermore, hypothalamic gene expression was affected to a greater extent in males than in females and, following EC exposure, male foetuses expressed more 'female-like' mRNA levels for some key neuroendocrine genes. This is the first study to show that 'real-life' maternal exposure to low levels of a complex cocktail of chemicals prior to conception can subsequently affect the developing foetal neuroendocrine system. These findings demonstrate that the developing neuroendocrine system is sensitive to EC mixtures in a sex-dimorphic manner likely to predispose to reproductive dysfunction in later life.


Assuntos
Disruptores Endócrinos/toxicidade , Exposição Materna , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/embriologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Caracteres Sexuais , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Masculino , Sistemas Neurossecretores/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Gravidez , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Kisspeptina-1/metabolismo , Receptores LHRH/metabolismo , Ovinos , Fatores de Tempo
17.
Trends Endocrinol Metab ; 9(9): 371-7, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18406308

RESUMO

Roles for oestrogens in brain masculinization/sexual behaviour, regulation of follicle-stimulating hormone (FSH)secretion and Leydig cell development and function are well established. However, the widespread distribution of oestrogen receptors alpha and beta in reproductive and other tissues of the male, and findings from human males or transgenic animals in which the genes coding for these receptors or for aromatase are non-functional, are changing our perception of the roles of oestrogen in the male. Aspects of pubertal development in boys (growth of the long bones, their mineralization and epiphyseal closure) attributed to the actions of androgens are now recognized as being mediated in part by oestrogens. Oestrogens also play a role (probably vasodilatatory) in the cardiovascular system of the male. Within the reproductive system, oestrogens have been shown to play a role in the regulation of fluid resorption from the efferent ducts and appear to be important in the structural and functional development of the Wolffian/excurrent duct system, as well as that of the prostate; inappropriately low or high oestrogen exposure during development can cause permanent changes to these tissues, which may lead to disorders of spermatogenesis and infertility. Sertoli cells and certain germ cells in the testis are also targets for oestrogen action. Many other tissues (adipose, kidney, thymus/immune system, skin, gut and muscle) are oestrogen targets in the male. Based on these findings and the widespread distribution of aromatase, it is argued that many of the effects of oestrogens in the male might stem from its local production and action and, furthermore, that the balance in action between androgens and oestrogens might be of central importance at many oestrogen target sites.

18.
Endocrinology ; 126(3): 1541-50, 1990 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2307118

RESUMO

This study has determined the route of secretion of inhibin-alpha into blood by the rat testis during sexual maturation, and in adult animals in which Leydig cell steroidogenesis was stimulated with human CG (hCG) or suppressed with aminoglutethimide. In each rat, inhibin-alpha levels were measured in samples of testicular (TV), spermatic (SV), and peripheral (PV) venous blood plasma, and in testicular interstitial fluid (IF). The IF and TV plasma reflect inhibin-alpha secretion via the base of the Sertoli cell while that secreted via the apex of the Sertoli cell (which is resorbed from the rete testis) was determined from the difference between SV and TV levels of inhibin-alpha. During sexual maturation, inhibin-alpha levels in IF and all plasma samples declined from maximal values at 28 days of age to minimal values at 100 days of age, in contrast to testosterone levels which showed the reverse pattern. There was a major change with age in the route of secretion of inhibin-alpha from the testis into blood. In immature (28-35 days) rats, most inhibin-alpha (58-65%) leaving the testis in blood was derived from that secreted via the base of the Sertoli cell with a relatively small contribution (35-42%) from apically-secreted inhibin-alpha. However, the latter made a progressively increasing contribution between 45 and 100 days of age (adults) and in adult rats the vast majority of inhibin-alpha (95%) leaving the testis in blood was derived from apically-secreted inhibin-alpha. This change was due primarily to a progressive reduction with age in the secretion of inhibin-alpha via the base of the Sertoli cell, a change which was confirmed by inhibin bioassay. Stimulation of steroidogenesis in the adult testis with hCG significantly increased inhibin-alpha and testosterone levels in IF and all plasma samples. The concomitant administration of hCG and aminoglutethimide (to block steroidogenesis) prevented the hCG-induced increase in testosterone levels, but still led to significant increases in inhibin-alpha secretion which were comparable to those seen with the use of hCG alone. The administration of aminoglutethimide (AMG) on its own did not alter the inhibin-alpha secretion profile from that seen in controls, but it did significantly reduce the levels of testosterone in all fluids. In rats treated with hCG +/- AMG there was a small change in the route of secretion of inhibin-alpha into blood, with an increased contribution (24-37%) from inhibin-alpha secreted via the base of the Sertoli cell, when compared with controls (7-16%).(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Inibinas/metabolismo , Maturidade Sexual/fisiologia , Esteroides/biossíntese , Testículo/metabolismo , Aminoglutetimida/farmacologia , Animais , Gonadotropina Coriônica/farmacologia , Células Intersticiais do Testículo/metabolismo , Masculino , Ratos , Ratos Endogâmicos , Esteroides/antagonistas & inibidores , Testosterona/metabolismo
19.
Endocrinology ; 116(6): 2592-604, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3922745

RESUMO

The effects of FSH on the testes of immature hypophysectomized rats were investigated by comparing functional changes in Leydig cells with changes in their number and morphological appearance. Rats were treated twice daily for 7 days with 0.5 ml saline vehicle, 10 micrograms rat FSH, or 20 ng ovine LH (an equivalent amount of LH known to contaminate the FSH preparation). FSH treatment caused a significant increase in testis weight and stimulated more advanced spermatogenic activity compared to saline or LH treatment. Morphometric analysis of glutaraldehyde perfusion-fixed testes showed no significant increase in Leydig cell number after LH treatment [saline, 4.63 +/- 0.14 million cells; LH, 6.38 +/- 0.55 million mean +/- SE)], but a significant (P less than 0.001) increase after FSH treatment (11.55 +/- 0.79 million). FSH, but not LH or saline, treatment resulted in Leydig cell hypertrophy and ultrastructural features identical to those of adult Leydig cells, these changes being reflected by enhanced hCG- and LHRH agonist-stimulated testosterone production in vitro by whole testes or dispersed interstitial cells. FSH and LH treatment caused minor but significant decreases in LH receptor numbers on dispersed interstitial cells compared to saline treatment. LHRH receptor numbers on interstitial cells were significantly increased only by FSH treatment. It is suggested that since FSH acts only on the seminiferous epithelium, then the hypertrophy, hyperplasia, and functional enhancement of Leydig cells after FSH treatment may be mediated by the secretion of one or more factors from the seminiferous tubules, providing additional evidence to support the view that gonadotropic regulation of testicular function is modulated by local interactions between the seminiferous tubules and the interstitial tissue.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Células Intersticiais do Testículo/fisiologia , Animais , Contagem de Células , Gonadotropina Coriônica/farmacologia , Hormônio Foliculoestimulante/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Técnicas In Vitro , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/farmacologia , Masculino , Ratos , Ratos Endogâmicos , Testosterona/biossíntese
20.
Endocrinology ; 128(1): 467-76, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1898883

RESUMO

This study has assessed whether depletion of specific germ cell types is able to alter the secretion of immunoactive inhibin by adult Sertoli cells in vivo and in vitro. Pachytene and later spermatocytes were depleted (80-100%) by a single administration of methoxy acetic acid (MAA; 650 mg/kg) to adult rats. At intervals between 1 and 42 days posttreatment, rats were killed, and the blood levels of FSH, LH, and testosterone were determined together with the levels of immunoactive inhibin in plasma and testicular interstitial fluid (IF). At the same time intervals, seminiferous tubules (ST; 5 x 2 cm) were isolated from control and MAA-treated rats and cultured for 24-72 h in the presence or absence of rat FSH, (Bu)2cAMP, or MAA under rigorously optimized conditions. The hormonal changes observed were related to the presence/absence of specific germ cell types, as determined by assessment of testicular morphology in perfusion-fixed testes from similarly treated rats. One to 3 days after MAA treatment, coincident with the depletion of pachytene spermatocytes, blood levels of FSH were increased significantly compared with controls; FSH returned to control levels at 7-14 days (when early spermatids were depleted), but were increased again at 21-35 days (when late spermatids were depleted). In contrast, while the plasma levels of immunoactive inhibin were increased 2-fold 3 days posttreatment, they were comparable to controls at 7-14 days, but were decreased substantially at 21-28 days. The levels of immunoactive inhibin in testicular IF were more than doubled 1 and 3 days posttreatment, but were comparable to control levels at all other times. Blood levels of LH showed a similar pattern of change to FSH, although only at 21-28 days after MAA treatment was there a significant increase, while blood levels of testosterone were comparable at all times in control and MAA-treated rats. To confirm that the changes observed in vivo after MAA treatment were indicative of changes in Sertoli rather than Leydig cell secretion of immonoactive inhibin, its secretion by isolated ST was assessed, and a pattern of change similar to that in plasma was observed. Thus, when cultured for 24 h under basal conditions, ST from rats 1-3 days after MAA treatment showed a 2- to 3-fold increase in secretion of immunoactive inhibin, which returned to control levels at 7-14 days before being reduced substantially at 21-28 days and then recovering to control levels; similar changes were observed for FSH- and (Bu)2cAMP-stimulated secretion of immunoactive inhibin.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Inibinas/metabolismo , Túbulos Seminíferos/metabolismo , Espermatozoides/fisiologia , Testículo/fisiologia , Acetatos/farmacologia , Animais , Hormônio Foliculoestimulante/sangue , Inibinas/sangue , Inibinas/imunologia , Hormônio Luteinizante/sangue , Masculino , Técnicas de Cultura de Órgãos , Radioimunoensaio , Ratos , Ratos Endogâmicos , Valores de Referência , Espermatócitos/efeitos dos fármacos , Espermatócitos/fisiologia , Espermatozoides/citologia , Testículo/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA