Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Reprod Fertil Dev ; 29(3): 609-620, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26488911

RESUMO

A dynamic partnership between follicle-stimulating hormone (FSH) and activin is required for normal Sertoli cell development and fertility. Disruptions to this partnership trigger Sertoli cells to deviate from their normal developmental pathway, as observed in inhibin α-knockout (Inha-KO) mice, which feature Sertoli cell tumours in adulthood. Here, we identified the developmental windows by which adult Sertoli cell tumourigenesis is most FSH sensitive. FSH was suppressed for 7 days in Inha-KO mice and wild-type littermates during the 1st, 2nd or 4th week after birth and culled in the 5th week to assess the effect on adult Sertoli cell development. Tumour growth was profoundly reduced in adult Inha-KO mice in response to FSH suppression during Weeks 1 and 2, but not Week 4. Proliferative Sertoli cells were markedly reduced in adult Inha-KO mice following FSH suppression during Weeks 1, 2 or 4, resulting in levels similar to those in wild-type mice, with greatest effect observed at the 2 week time point. Apoptotic Sertoli cells increased in adult Inha-KO mice after FSH suppression during Week 4. In conclusion, acute FSH suppression during the 1st or 2nd week after birth in Inha-KO mice profoundly suppresses Sertoli cell tumour progression, probably by inhibiting proliferation in the adult, with early postnatal Sertoli cells being most sensitive to FSH action.


Assuntos
Inibinas/metabolismo , Tumor de Células de Sertoli/patologia , Espermatogênese/genética , Neoplasias Testiculares/patologia , Ativinas/sangue , Animais , Hormônio Foliculoestimulante/sangue , Inibinas/genética , Masculino , Camundongos , Camundongos Knockout , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Testículo/metabolismo , Testículo/patologia
2.
Reprod Fertil Dev ; 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26946412

RESUMO

Although oestrogens are essential for spermatogenesis and their biosynthesis is dependent on aromatase expression, the molecular mechanism of aromatase regulation is poorly understood. Our laboratory has demonstrated that liver kinase B1 (LKB1) is a negative regulator of aromatase in the breast by phosphorylating AMP-activated protein kinase (AMPK) and inhibiting the nuclear translocation of the cAMP response element-binding protein-regulated transcription co-activator (CRTC) 2. The aim of this study was to determine the location of testis-associated proteins in the LKB1-CRTC pathway. Aromatase, LKB1, phosphorylated AMPK (pAMPK) and CRTC1-3 were examined by selected immunofluorescent antibodies in testis samples from a prepubertal boy and three fertile men. Aromatase, pAMPK and LKB1 proteins were present in the seminiferous epithelium and interstitium of the testis and were expressed in a differential and developmental manner in particular cell types. The expression pattern of LKB1 was similar to that of pAMPK and inversely related to aromatase expression. CRTC1 and CRTC3 were localised in the seminiferous epithelium, whereas CRTC2 was barely detectable in testis. These results lead to the conclusion that LKB1 is involved in the molecular pathway that underpins aromatase regulation in the testis via CRTC1 and CRTC3 and may be important for the oestrogen-mediated development of germ cells.

3.
Reprod Fertil Dev ; 2014 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-25515817

RESUMO

We tested whether the reversible effects of nutrition on spermatogenesis in sexually mature sheep were mediated by Sertoli cells. Rams were fed with diets designed to achieve a 10% increase (High), no change (Maintenance) or a 10% decrease (Low) in body mass after 65 days. At the end of treatment, testes were lighter in the Low than the High group (PP<0.05) in the expression of seven Sertoli cell-specific genes. Under-nutrition appeared to reverse cellular differentiation leading to disruption of tight-junction morphology. In conclusion, in sexually mature sheep, reversible reductions in testis mass and spermatogenesis caused by under-nutrition were associated with impairment of basic aspects of Sertoli cell function but not with changes in the number of Sertoli cells.

4.
Biol Reprod ; 87(1): 13, 1-11, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22492971

RESUMO

New data have challenged the convention that the adult Sertoli cell population is fixed and unmodifiable. The Sertoli cell has two distinct functions: 1) formation of the seminiferous cords and 2) provision of nutritional and structural support to developing germ cells. For these to occur successfully, Sertoli cells must undergo many maturational changes between fetal and adult life, the main switches occurring around puberty, including the loss of proliferative activity and the formation of the blood-testis barrier. Follicle-stimulating hormone plays a key role in promoting Sertoli cell proliferation, while thyroid hormone inhibits proliferative activity in early postnatal life. Together these regulate the Sertoli-germ cell complement and sperm output in adulthood. By puberty, the Sertoli cell population is considered to be stable and unmodifiable by hormones. But there is mounting evidence that the size of the adult Sertoli cell population and its maturational status is modifiable by hormones and that Sertoli cells can gain proliferative ability in the spermatogenically disrupted hamster and human model. This new information demonstrates that the adult Sertoli cell population, at least in the settings of testicular regression in the hamster and impaired fertility in humans in vivo and from mice and men in vitro, is not a terminally differentiated population. Data from the hamster now show that the adult Sertoli cell population size is regulated by hormones. This creates exciting prospects for basic and clinical research in testis biology. The potential to replenish an adult Sertoli-germ cell complement to normal in a setting of infertility may now be realized.


Assuntos
Células de Sertoli/citologia , Animais , Comunicação Autócrina/fisiologia , Proteínas de Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Humanos , Junções Intercelulares/fisiologia , Masculino , Modelos Biológicos , Comunicação Parácrina/fisiologia , Puberdade/fisiologia , Estações do Ano , Células de Sertoli/fisiologia , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia , Espermatogênese/fisiologia
5.
Biol Reprod ; 84(2): 379-91, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20926807

RESUMO

Activin affects many aspects of cellular development, including those essential for reproductive fitness. This study examined the contribution of activin A to murine fetal testicular development, revealing contrasting outcomes of activin actions on Sertoli cells and gonocytes. Shortly after sex determination, from Embryonic Day 12.5 (E12.5) through to birth (0 dpp), the activin A subunit transcript (Inhba) level rises in testis but not ovary, followed closely by the Inha transcript (encoding the inhibitory inhibin alpha subunit). Activin receptor transcript levels also change, with Acvr1 (encoding ALK2) and Acvr2b (ActRIIB) significantly higher and lower, respectively, at 0 dpp compared with E13.5 and E15.5. Transcripts encoding the signaling mediators Smad1, Smad3, and Smad4 were higher at 0 dpp compared with E13.5 and E15.5, whereas Smad2, Smad5, and Smad7 were lower. Detection of phosphorylated (P-)SMAD2/3 in nearly all testis cell nuclei indicated widespread transforming growth factor beta (TGFB) and/or activin ligand signaling activity. In contrast to wild-type littermates, activin betaA subunit knockout (Inhba(-/-)) mice have significantly smaller testes at birth, attributable to a 50% lower Sertoli cell number and decreased Sertoli cell proliferation from E13.5. Inhba(-/-) testes contained twice the normal gonocyte number at birth, with some appearing to bypass quiescence. Persistence of widespread P-SMAD2/3 in Inhba(-/-) cells indicates other TGFB superfamily ligands are active in fetal testes. Significant differences in Smad and cell cycle regulator transcript levels correlating to Inhba gene dosage correspond to differences in Sertoli and germ cell numbers. In Inhba(-/-) testes, Cdkn1a (encoding p21(cip1)), identified previously in fetal gonocytes, was lower at E13.5, whereas Cdkn1b (encoding p(27kip1) in somatic cells) was lower at birth, and cyclin D2 mRNA and protein were lower at E15.5 and 0 dpp. Thus, activin A dosage contributes to establishing the balance between Sertoli and germ cell number that is ultimately required for adult male fertility.


Assuntos
Ativinas/metabolismo , Proliferação de Células , Feto/citologia , Subunidades beta de Inibinas/metabolismo , Células de Sertoli/citologia , Espermatozoides/citologia , Testículo/embriologia , Receptores de Ativinas/genética , Animais , Contagem de Células , Ciclo Celular/fisiologia , Feminino , Fertilidade/fisiologia , Desenvolvimento Fetal , Feto/anatomia & histologia , Feto/metabolismo , Idade Gestacional , Subunidades beta de Inibinas/genética , Masculino , Camundongos , Tamanho do Órgão , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo
6.
Biol Reprod ; 82(5): 980-90, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20130270

RESUMO

The transforming growth factor beta superfamily ligand activin A controls juvenile testis growth by stimulating Sertoli cell proliferation. Testicular levels are highest in the first postnatal week, when Sertoli cells are proliferating and spermatogonial stem cells first form. Levels decrease sharply as Sertoli cell proliferation ceases and spermatogenic differentiation begins. We hypothesized that changing activin levels also affect germ cell maturation. We detected an acute and developmentally regulated impact of activin on Kit mRNA in cocultures of Sertoli cells and germ cells from Day 8, but not Day 4, mice. Both stereological and flow cytometry analyses identified an elevated spermatogonium:Sertoli cell ratio in Day 7 testes from Inhba(BK/BK) mice, which have decreased bioactive activin, and the germ cell markers Sycp3, Dazl, and Ccnd3 were significantly elevated in Inhba(BK/BK) mice. The flow cytometry measurements demonstrated that surface KIT protein is significantly higher in Day 7 Inhba(BK/BK) germ cells than in wild-type littermates. By Day 14, the germ cell:Sertoli cell ratio did not differ between genotypes, but the transition of type A spermatogonia into spermatocytes was altered in Inhba(BK/BK) testes. We conclude that regulated activin signaling not only controls Sertoli cell proliferation, as previously described, but also influences the in vivo progression of germ cell maturation in the juvenile testis at the onset of spermatogenesis.


Assuntos
Subunidades beta de Inibinas/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Animais , Diferenciação Celular , Técnicas de Cocultura , Células Germinativas/metabolismo , Subunidades beta de Inibinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , RNA Mensageiro/análise , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Células de Sertoli/citologia , Maturação do Esperma , Espermatogênese/fisiologia , Espermatogônias/citologia , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Testículo/citologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
7.
Int J Androl ; 32(5): 542-55, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18522674

RESUMO

Celecoxib (Celebrex), an inhibitor of cyclooxygenase-2 (COX-2; prostaglandin-endoperoxide synthase 2; EC 1.14.99.1), is widely used in the treatment of chronic inflammation and pain. COX-2 is constitutively expressed in the testis, where it is responsible for prostaglandin production, so inhibition of this enzyme should have effects on testicular function. The effects of administering celecoxib (oral with feed, 0.15% w/w) for 5 weeks on normal testis function and the response to low dose (0.1 mg/kg body weight) or high dose (5.0 mg/kg) lipopolysaccharide (LPS) were examined in adult male rats. Celecoxib caused a 60% reduction in testicular interstitial fluid (IF) prostaglandin E(2) (PGE(2)) concentrations, accompanied by a compensatory increase in COX-2 mRNA expression. Celecoxib increased IF volume by 30%, but had no effect on testis weight, testis morphology or serum testosterone levels. In the celecoxib-fed rats, the dose-dependent inhibitory effects of LPS on testis weight, IF volume and serum testosterone levels were significantly diminished. However, celecoxib had no effect on COX-2 protein levels or LPS-induced expression of the inflammatory mediators interleukin-1beta, tumour necrosis factor-alpha or inducible nitric-oxide synthase. A similar lack of inhibition of LPS-induced cytokine expression by another COX-2 inhibitor, NS-398, was observed in vitro. These data indicate that celecoxib reduces intratesticular activity of COX-2 (as indicated by PGE(2) levels) and inhibits IF formation in the testis, but has no appreciable effect on steroidogenesis or spermatogenesis, at least in the short term. Celecoxib does not appear to alter the ability of the testis to mount an inflammatory response but opposes the deleterious effects of inflammation on IF formation and testosterone production. These results indicate significant roles for products of the COX-2 pathway in testicular vascular control and steroidogenesis, which may have implications for men with marginal fertility taking celecoxib for extended periods, but also highlight the potential of this drug to ameliorate testicular damage caused by systemic or local inflammation.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Lipopolissacarídeos/farmacologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Testículo/efeitos dos fármacos , Animais , Sequência de Bases , Celecoxib , Primers do DNA , Dinoprostona/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/patologia , Testículo/fisiologia
8.
Hum Reprod ; 23(2): 403-11, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18199736

RESUMO

BACKGROUND: Gonadotrophins support spermatogenesis via poorly understood mechanisms. We aimed to determine the effect of FSH/LH suppression in regulating germ cell apoptosis and proliferation in normal fertile men. METHODS: Testicular tissues were obtained after gonadotrophin suppression induced by testosterone alone or combined with depot medroxyprogesterone acetate for 2 or 6 weeks and an untreated group of men (referred to as 'normal men') served as controls (n = 5 or 10 men per group). Apoptosis and proliferation were identified by terminal deoxynucleotidyl transferase-mediated dUDP nick-end labelling (TUNEL) and proliferating cell nuclear antigen (PCNA) labelling methods, respectively. Intrinsic and extrinsic apoptotic pathways were identified by immunohistochemistry using the pathway-specific proteins: activated caspase (aCaspase) 9 and 8 and quantified using stereological techniques. RESULTS: By 2 and 6 weeks, the proportion of TUNEL-labelled spermatogonia increased to 354% and 268% respectively, compared with normal men (P < 0.001), with increased caspase 9 [223 and 166% compared with normal men (P < 0.001)], but no increase in caspase 8, immunoreactivity. At 6 weeks, the proportions of TUNEL-labelled spermatocytes and round spermatids tended to increase (303 and 180% compared with normal men, NS), as did caspase 9 (199 and 147% compared with normal men, NS) and caspase 8 immunoreactivities (286 and 243% compared with normal men, NS and P = 0.06), respectively. The proportion of TUNEL-labelled elongating/elongated spermatids tended to increase (144 and 138% compared with normal men, NS) at 2 and 6 weeks, respectively, with no change in either caspase immunoreactivities. Even though the number of PCNA-labelled cells did not change with gonadotrophin suppression, the balance between proliferation and apoptosis was lower in spermatogonia (P = 0.01) and spermatocytes (P = 0.3) between treated and untreated normal men. CONCLUSIONS: We demonstrated that gonadotrophins act as spermatogonial survival factors via the regulation of intrinsic apoptotic pathway, whereas having no effect of cellular proliferation in normal men.


Assuntos
Gonadotropinas/fisiologia , Espermatogônias/fisiologia , Adulto , Apoptose/fisiologia , Caspase 8/metabolismo , Caspase 9/metabolismo , Forma Celular , Sobrevivência Celular/fisiologia , Combinação de Medicamentos , Ativação Enzimática , Gonadotropinas/antagonistas & inibidores , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Acetato de Medroxiprogesterona/farmacologia , Pessoa de Meia-Idade , Valores de Referência , Espermátides/citologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Espermatogônias/enzimologia , Testosterona/farmacologia
9.
Reproduction ; 135(6): 867-77, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18502899

RESUMO

This study aimed to assess the effect of gonadotrophin suppression and FSH replacement on testicular tight junction dynamics and blood-testis barrier (BTB) organisation in vivo, utilising the seasonal breeding Djungarian hamster. Confocal immunohistology was used to assess the cellular organisation of tight junction proteins and real-time PCR to quantify tight junction mRNA. The effect of tight junction protein organisation on the BTB permeability was also investigated using a biotin-linked tracer. Tight junction protein (claudin-3, junctional adhesion molecule (JAM)-A and occludin) localisation was present but disorganised after gonadotrophin suppression, while mRNA levels (claudin-11, claudin-3 and occludin) were significantly (two- to threefold) increased. By contrast, both protein localisation and mRNA levels for the adaptor protein zona occludens-1 decreased after gonadotrophin suppression. FSH replacement induced a rapid reorganisation of tight junction protein localisation. The functionality of the BTB (as inferred by biotin tracer permeation) was found to be strongly associated with the organisation and localisation of claudin-11. Surprisingly, JAM-A was also recognised on spermatogonia, suggesting an additional novel role for this protein in trans-epithelial migration of germ cells across the BTB. It is concluded that gonadotrophin regulation of tight junction proteins forming the BTB occurs primarily at the level of protein organisation and not gene transcription in this species, and that immunolocalisation of the organised tight junction protein claudin-11 correlates with BTB functionality.


Assuntos
Barreira Hematotesticular/metabolismo , Hormônio Foliculoestimulante/fisiologia , Proteínas de Membrana/metabolismo , Phodopus/metabolismo , Testículo/metabolismo , Junções Íntimas/metabolismo , Animais , Sequência de Bases , Barreira Hematotesticular/efeitos dos fármacos , Moléculas de Adesão Celular/análise , Moléculas de Adesão Celular/genética , Claudina-3 , Claudinas , Cricetinae , Primers do DNA/genética , Imunofluorescência , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica/efeitos dos fármacos , Moléculas de Adesão Juncional , Masculino , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Microscopia Confocal , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Ocludina , Fosfoproteínas/análise , Fosfoproteínas/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Estações do Ano , Transcrição Gênica , Proteína da Zônula de Oclusão-1
10.
J Endocrinol ; 192(3): 553-61, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17332524

RESUMO

The roles of testosterone (T) and its metabolites on hamster spermatogenesis are poorly defined. This study assessed the effects of T, dihydrotestosterone (DHT) and oestradiol (E) on the re-initiation of spermatogenesis in the adult Djungarian hamster. Hamsters raised under long photoperiods (LD, 16 h light:8 h darkness) were exposed to short photoperiods (SD, 8 h light:16 h darkness) for 11 weeks to suppress gonadotrophins. Groups of eight animals then received T, DHT and E for 5 weeks. Cell numbers were determined using the optical disector (sic). The number of Sertoli cells was suppressed in SD controls to 48% (P < 0.001) of LD control and restored either fully or partially by exogenous DHTand E (2.6- and 1.8-fold above SD levels) respectively, corresponding with a twofold elevation of serum FSH. The number of germ cells in SD animals was reduced (all P < 0.001) to levels reported. The number of type A spermatogonia increased in line with the rise in Sertoli cell number, by 2.6-fold (P < 0.01) and 1.8-fold (NS) above SD controls after DHT and E treatments respectively. DHT increased the number of type B spermatogonia/preleptotene spermatocytes, leptotene/zygotene and pachytene spermatocytes by 3.5-, 5.7- and 21-fold above SD (all P < 0.01) respectively, compared with a 2.2-fold (P < 0.01), 2.4-fold (not significant, NS) and 6-fold (NS) in E-treated animals respectively. Exogenous T had little effect on cell numbers or serum FSH compared with SD controls. Spermatids were rarely observed after steroid treatment. We believe this study suggests that steroids can regulate the re-initiation of early spermatogenic cells via a mechanism which includes FSH.


Assuntos
Hormônios Esteroides Gonadais/farmacologia , Phodopus/fisiologia , Fotoperíodo , Espermatogênese/efeitos dos fármacos , Testículo/fisiologia , Animais , Contagem de Células , Cricetinae , Di-Hidrotestosterona/farmacologia , Implantes de Medicamento , Estradiol/farmacologia , Hormônio Foliculoestimulante/sangue , Masculino , Radioimunoensaio , Células de Sertoli/fisiologia , Espermatócitos/citologia , Espermatogônias/citologia , Testículo/efeitos dos fármacos , Testosterona/farmacologia
11.
J Androl ; 28(1): 176-85, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16988326

RESUMO

Male fertility is inhibited by inflammatory disease, but the mechanisms responsible are poorly defined. The effects of acute systemic inflammation induced by a single IP injection of lipopolysaccharide (LPS) on spermatogenic function in adult male rats were investigated using detailed stereological analysis. The earliest effect observed was a significant maturational delay of meiosis during the leptotene/zygotene phase (at stages IX-XIII) within 24 hours. This was followed within 6 days by an increase in premature release of these cells and the adjacent, more luminally located generation of round spermatids from the seminiferous epithelium. An increase in germ cell apoptosis within stages IX-XIII also occurred at this time. These data indicate that the initial effects of acute inflammation on the seminiferous epithelium are most pronounced on stages IX-XIII. The effects were not consistent with a loss of hormonal regulation, suggesting that a direct effect of inflammation on the function of the Sertoli cell during this critical stage of meiosis is involved. In the longer term, however, the consequences of this acute inflammatory episode were relatively minor: within 28 days there had been a compensatory increase in the efficiency of the seminiferous epithelium, restoring the spermatogenic capacity of the testis towards preinflammation levels.


Assuntos
Inflamação/fisiopatologia , Espermatogênese , Animais , Apoptose , Contagem de Células , Hormônios Esteroides Gonadais/metabolismo , Inflamação/induzido quimicamente , Lipopolissacarídeos , Masculino , Ratos , Ratos Sprague-Dawley , Testículo/patologia
12.
Endocrinology ; 147(1): 472-82, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210366

RESUMO

Simultaneous suppression of both testosterone and FSH with GnRH antagonists (GnRH-ant) reverses the radiation-induced block in spermatogonial differentiation in F1 hybrids of Lewis and Brown-Norway rats. Although addition of exogenous testosterone restores the block, it also raises FSH, and hence it had not been possible to conclusively determine which hormone was inhibiting spermatogonial differentiation. In the present study, we establish the relative roles of testosterone and FSH in this inhibition using three different approaches. The first approach involved the treatment of irradiated rats, in which differentiation was stimulated by GnRH-ant plus flutamide, with FSH for 2 wk; the FSH reduced the percentage of tubules that were differentiated (TDI) by about 2-fold, indicating that FSH does have an inhibitory role. The second approach involved treatment of irradiated, hypophysectomized rats with exogenous testosterone for 10 wk; testosterone also reduced the TDI, demonstrating that testosterone had a definite inhibitory effect, independent of pituitary hormones. Furthermore, in this protocol we showed that TDI in the hypophysectomized testosterone-treated group, which had higher intratesticular testosterone levels but lacked FSH, was slightly higher than the TDI in a GnRH-antagonist-testosterone-treated group of irradiated rats, which had normal physiological levels of FSH; this result supports a role for endogenous FSH in suppressing spermatogonial differentiation in the latter group. The third approach involved injection of an active anti-FSH antibody for 10 d in untreated, GnRH-ant plus flutamide-treated, or GnRH-ant plus testosterone-treated irradiated rats. This was not sufficient to increase the TDI. However, flutamide given in a similar treatment schedule did increase the TDI in GnRH-ant plus testosterone-treated rats. We conclude that both testosterone and FSH individually inhibit spermatogonial differentiation after irradiation, but testosterone is a more highly potent inhibitor than is FSH.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Espermatogônias/citologia , Espermatogônias/efeitos da radiação , Testosterona/farmacologia , Animais , Flutamida/farmacologia , Humanos , Hipofisectomia , Masculino , Ratos , Proteínas Recombinantes/farmacologia , Espermatogônias/efeitos dos fármacos
13.
J Clin Endocrinol Metab ; 91(10): 3962-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16895950

RESUMO

CONTEXT: Male hormonal contraception via gonadotropin and intratesticular androgen withdrawal disrupts spermatogenesis at two principal sites: 1) spermatogonial maturation, and 2) spermiation. OBJECTIVE: The objective of this study was to explore the relative dependence of each stage of germ cell development on FSH and LH/intratesticular androgen action. DESIGN, SETTING, AND PARTICIPANTS: Eighteen men enrolled in this prospective, randomized 14-wk study at Prince Henry's Institute. INTERVENTIONS: Subjects (n = 6/group) were assigned to 6 wk of 1) testosterone (T) implant (4 x 200 mg sc once)+depot medroxy progesterone acetate (DMPA; 150 mg im once); 2) T implant+DMPA+FSH (300 IU sc twice weekly); and 3) T implant+DMPA+human chorionic gonadotropin (hCG; 1000 IU sc twice weekly as an LH substitute). Men then underwent a vasectomy and testicular biopsy with previously reported control data used for comparison. MAIN OUTCOME MEASURES: Germ cell number (assessed by the optical disector stereological approach) and intratesticular androgen levels were determined. RESULTS: T+DMPA alone significantly suppressed type B spermatogonia, preleptotene through to pachytene spermatocytes, and round spermatids from control (P < 0.05). All germ cell subtypes were maintained at control levels by either FSH or LH activity, except pachytene spermatocytes, which were found to be lower in the hCG vs. FSH (P < 0.01) and control groups (P < 0.05). CONCLUSIONS: FSH and LH maintained spermatogenesis independently in this gonadotropin-suppressed model. Compared with LH, FSH showed better maintenance of pachytene spermatocyte number, whereas improved conversion to round spermatids was suggested with hCG treatment. Future contraceptive treatment strategies must consider independent regulation of spermatogenesis by both FSH and LH/intratesticular androgens for maximum efficacy.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Hormônio Luteinizante/fisiologia , Espermatogênese , Espermatogônias/fisiologia , Adulto , Gonadotropina Coriônica/farmacologia , Estradiol/sangue , Hormônio Foliculoestimulante/sangue , Humanos , Hormônio Luteinizante/sangue , Masculino , Acetato de Medroxiprogesterona/administração & dosagem , Pessoa de Meia-Idade , Estudos Prospectivos , Contagem de Espermatozoides , Testículo/química , Testosterona/administração & dosagem , Testosterona/sangue
14.
J Clin Endocrinol Metab ; 90(10): 5647-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16030154

RESUMO

CONTEXT: Combination of a GnRH antagonist (acyline), types I and II, 5alpha-reductase inhibitor (dutasteride) or levonorgestrel (LNG) with testosterone (T) treatment may augment the suppression of spermatogenesis and intratesticular (iT) steroids. OBJECTIVE: The objective of this study was to assess the effects of combined hormonal contraceptive regimens on germ cell populations and iT steroids. DESIGN, SETTING, AND PARTICIPANTS: Twenty-nine normal health men enrolled in this prospective, randomized, 14-wk study at the University of Washington. INTERVENTION(S): Twenty-two men (n = 5-6/group) received 8 wk of T enanthate (TE; 100 mg, i.m., weekly) combined with 1) 125 microg LNG daily, orally; 2) 125 microg LNG plus 0.5 mg dutasteride daily, orally; 3) 300 microg/kg acyline twice weekly, s.c.; or 4) 125 microg LNG daily, orally, plus 300 microg/kg acyline twice weekly, s.c. Subjects then underwent a vasectomy and testicular biopsy. Control men (n = 7) proceeded directly to surgery. MAIN OUTCOME MEASURE(S): The main outcome measures were germ cells and iT steroids [T, dihydrotestosterone, 3alpha- and beta-androstanediol (Adiol), and estradiol (E2)]. RESULTS: High iT levels of all androgens (6- to 123-fold serum levels) and E2 (407-fold serum levels) were found in control men. iTT (1.9-2.6% control; P < 0.001) and iT3betaAdiol (16-34% control; P < 0.05) levels decreased with all treatments. iT dihydrotestosterone (13-29% control; P < 0.05) and iT3alphaAdiol (44-47% control; P < 0.05) levels decreased with all but the TE plus LNG treatment. iTE2 levels decreased only in the TE plus acyline group (28% control; P = 0.01). Germ cells from type B spermatogonia onward were suppressed, with no differences between groups found. Variable sites of impairment of germ cell progression were evident between men (spermagonial maturation, meiosis 1 entry, and spermiation). Other than a negative correlation between iT3alphaAdiol and haploid germ cell number (P < 0.006), no correlations between germ cell number and gonadotropins, sperm concentration, or iT steroids were found. CONCLUSIONS: A similar high testicular:serum gradient exists for E2 and T in normal men, and 8 wk of gonadotropin suppression markedly reduces iTT, with 5alpha-reduced androgens and E2 levels decreasing to a much lesser degree. The heterogeneity of the germ cell response, regardless of treatment, gonadotropins or iT steroids, points to the individual sensitivity of sites in germ cell development, which is worthy of additional exploration.


Assuntos
Inibidores de 5-alfa Redutase , Inibidores Enzimáticos/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Levanogestrel/farmacologia , Congêneres da Progesterona/farmacologia , Espermatogênese/efeitos dos fármacos , Esteroides/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testosterona/farmacologia , Adulto , Androstanos/sangue , Azasteroides/farmacologia , Di-Hidrotestosterona/sangue , Dutasterida , Estradiol/sangue , Células Germinativas/efeitos dos fármacos , Humanos , Masculino , Estudos Prospectivos , Células de Sertoli/efeitos dos fármacos , Testosterona/sangue , Vasectomia
15.
J Endocrinol ; 185(3): 529-38, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15930179

RESUMO

Oestrogen is a metabolite of testosterone, but its role in spermatogenesis is ill-defined. Oestrogen may exert its effects on spermatogenesis, as oestrogen receptor (ER)-beta has been localised to both germ and somatic cells. This study sought to establish whether the restoration of early germ cell numbers in spermatogenesis by high-dose exogenous testosterone was influenced by its metabolite, oestrogen. The ER antagonist (ICI 182780) was administered, at a dose known to impair oestrogen action in the male reproductive tract, during testosterone treatment of gonadotrophin-releasing hormone (GnRH)-immunised rats, and germ cell numbers were determined. GnRH-immunised adult Sprague-Dawley rats (n=7-8 per group) received two doses of testosterone, either as a Silastic implant (24 cm (T24 cm)) or an injectable ester for 10 days alone or in combination with ICI 182780 (2 mg/kg, s.c. injection daily). Control rats received vehicle alone. Testes were perfusion-fixed and germ cells were quantified by the optical disector technique. GnRH-immunisation reduced (P<0.001) both type A/ intermediate spermatogonial and type B spermatogonial/ preleptotene spermatocyte number (56% of control) and leptotene/zygotene spermatocyte number (63% of control). Pachytene spermatocyte and round spermatids were reduced to 12% and l% (P<0.01) of control respectively. Testosterone treatment did not increase type A/intermediate spermatogonial number compared with GnRH-immunised controls over the 10-day study period. Treatment with testosterone-esters increased type B spermatogonial/preleptotene spermatocytes and leptotene/zygotene spermatocyte numbers (both being ~83% of control, P<0.05), while T24 cm treatment did not significantly increase their numbers (~73% of control) compared with GnRH-immunised controls. Both treatments increased pachytene spermatocyte and round spermatid numbers to 55% and 8% of control respectively. Co-administration of ICI 182780 had no effect on any of these germ cell numbers. We conclude that oestrogen action plays no role in the short-term restoration of spermatogenesis by testosterone in the GnRH-immunised rat.


Assuntos
Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Espermatogênese/efeitos dos fármacos , Animais , Estrogênios/farmacologia , Hormônio Foliculoestimulante/farmacologia , Fulvestranto , Imunização , Hormônio Luteinizante/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Contagem de Espermatozoides , Espermátides/efeitos dos fármacos , Espermatócitos/efeitos dos fármacos , Testosterona/sangue , Testosterona/farmacologia
16.
J Endocrinol ; 186(3): 429-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16135663

RESUMO

The critical influence of follicle stimulating hormone (FSH) on male fertility relates both to its impact on Sertoli cell proliferation in perinatal life and to its influence on the synthesis of Sertoli cell-derived products essential for germ cell survival and function in the developing adult testis. The nature and timing of this shift of germ cells to their reliance on specific Sertoli cell-derived products are not defined. Based on existing data, it is apparent that the dominant function of FSH shifts between 9 and 18 day postpartum (dpp) during the first wave of spermatogenesis from driving Sertoli cell proliferation to support germ cells. To enable comprehensive analysis of the impact of acute in vivo FSH suppression on Sertoli and germ cell development, FSH was selectively suppressed in Sprague-Dawley rats by passive immunisation for 2 days and/or 4 days prior to testis collection at 3, 9 and 18 dpp. The 3 dpp samples displayed no measurable changes, while 4 days of FSH suppression decreased Sertoli cell proliferation and numbers in 9 dpp, but not 18 dpp, animals. In contrast, germ cell numbers were unaffected at 9 dpp but decreased at 18 dpp following FSH suppression, with a corresponding increase in germ cell apoptosis measured at 18 dpp. Sixty transcripts were measured as changed at 18 dpp in response to 4 days of FSH suppression, as assessed using Affymetrix microarrays. Some of these are known as Sertoli cell-derived FSH-responsive genes (e.g. StAR, cathepsin L, insulin-like growth factor binding protein-3), while others encode proteins involved in cell cycle and survival regulation (e.g. cyclin D1, scavenger receptor class B 1). These data demonstrate that FSH differentially affects Sertoli and germ cells in an age-dependent manner in vivo, promoting Sertoli cell mitosis at day 9, and supporting germ cell viability at day 18. This model has enabled identification of candidate genes that contribute to the FSH-mediated pathway by which Sertoli cells support germ cells.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Testículo/crescimento & desenvolvimento , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Primers do DNA , Hormônio Foliculoestimulante/imunologia , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Imunização Passiva , Inibinas/sangue , Masculino , Análise em Microsséries , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/patologia , Testículo/efeitos dos fármacos , Testosterona/sangue , Fatores de Tempo
17.
Endocrinology ; 155(3): 1131-44, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24424039

RESUMO

The blood-testis barrier (BTB) sequesters meiotic spermatocytes and differentiating spermatids away from the vascular environment. We aimed to assess whether meiosis and postmeiotic differentiation could occur when the BTB is permeable. Using a model of meiotic suppression and reinitiation, BTB function was assessed using permeability tracers of small, medium, and large (0.6-, 70-, and 150-kDa) sizes to emulate blood- and lymphatic-borne factors that could cross the BTB. Adult rats (n = 9/group) received the GnRH antagonist acyline (10 wk) to suppress gonadotropins, followed by testosterone (24cm Silastic implant), for 2, 4, 7, 10, 15, and 35 days. In acyline-suppressed testes, all tracers permeated the seminiferous epithelium. As spermatocytes up to diplotene stage XIII reappeared, both the 0.6- and 70-kDa tracers, but not 150 kDa, permeated around these cells. Intriguingly, the 0.6- and 70-kDa tracers were excluded from pachytene spermatocytes at stages VII and VIII but not in subsequent stages. The BTB became progressively impermeable to the 0.6- and 70-kDa tracers as stages IV-VII round spermatids reappeared in the epithelium. This coincided with the appearance of the tight junction protein, claudin-12, in Sertoli cells and at the BTB. We conclude that meiosis can occur when the BTB is permeable to factors up to 70 kDa during the reinitiation of spermatogenesis. Moreover, BTB closure corresponds with the presence of particular pachytene spermatocytes and round spermatids. This research has implications for understanding the effects of BTB dynamics in normal spermatogenesis and also potentially in states where spermatogenesis is suppressed, such as male hormonal contraception or infertility.


Assuntos
Barreira Hematotesticular/fisiologia , Espermatogênese , Animais , Claudinas/metabolismo , Hormônio Foliculoestimulante/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Microscopia de Fluorescência , Oligopeptídeos/metabolismo , Tamanho do Órgão , Permeabilidade , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/metabolismo , Espermátides/citologia , Espermatócitos/citologia , Testículo/metabolismo , Junções Íntimas , Fatores de Tempo
18.
Spermatogenesis ; 3(1): e24014, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23687617

RESUMO

It is widely held that the somatic cell population that is responsible for sperm development and output (Sertoli cells) is terminally differentiated and unmodifiable in adults. It is postulated, with little evidence, that Sertoli cells are not terminally differentiated in some phenotypes of infertility and testicular cancer. This study sought to compare markers of Sertoli cell differentiation in normospermic men, oligospermic men (undergoing gonadotropin suppression) and testicular carcinoma in situ (CIS) and seminoma samples. Confocal microscopy was used to assess the expression of markers of proliferation (PCNA and Ki67) and functional differentiation (androgen receptor). As additional markers of differentiation, the organization of Sertoli cell tight junction and associated proteins were assessed in specimens with carcinoma in situ. In normal men, Sertoli cells exhibited a differentiated phenotype (i.e., PCNA and Ki67 negative, androgen 40 receptor positive). However, after long-term gonadotropin suppression, 1.7 ± 0.6% of Sertoli cells exhibited PCNA reactivity associated with a diminished immunoreactivity in androgen receptor, suggesting an undifferentiated phenotype. Ki67-positive Sertoli cells were also observed. PCNA-positive Sertoli cells were never observed in tubules with carcinoma in situ, and only rarely observed adjacent to seminoma. Tight junction protein localization (claudin 11, JAM-A and ZO-1) was altered in CIS, with a reduction in JAM-A reactivity in Sertoli cells from tubules with CIS and the emergence of strong JAM-A reactivity in seminoma. These findings indicate that adult human Sertoli cells exhibit characteristics of an undifferentiated state in oligospermic men and patients with CIS and seminoma in the presence of germ cell neoplasia.

19.
J Clin Endocrinol Metab ; 98(12): E1979-87, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24037887

RESUMO

CONTEXT: Peutz-Jeghers syndrome (PJS) is an autosomal-dominant disorder that arises as a consequence of mutations in the STK11 gene that encodes LKB1. PJS males often have estrogen excess manifesting as gynecomastia and advanced bone age. We and others have previously described an increase in testicular aromatase expression in PJS patients. However, the underlying mechanism has not yet been explored. OBJECTIVE: The aim of this study was to characterize the role of LKB1 in regulating the expression of aromatase in boys with PJS via signaling pathways involving AMP-activated protein kinase (AMPK) and cyclic AMP-responsive element binding protein-regulated transcription coactivators (CRTCs). PATIENTS: We studied testicular biopsies from two boys with STK11 mutations: a 13-year-old boy and an unrelated 4-year-old boy with prepubertal gynecomastia and advanced bone age, as well as breast tissue from the 13-year-old boy. RESULTS: Loss of heterozygosity of STK11, measured by the absence of LKB1 immunofluorescence, was observed in Sertoli cells of abnormal cords of testis samples from affected individuals. This was associated with loss of p21 expression and decreased phosphorylation of AMPK, known downstream targets of LKB1, as well as the increased expression of aromatase. Similar results of low LKB1 expression in cells expressing aromatase were observed in the mammary epithelium from one of these individuals. Nuclear expression of the CRTC proteins, potent stimulators of aromatase and known to be inhibited by AMPK, was significantly correlated with aromatase. CONCLUSIONS: Loss of heterozygosity of the STK11 gene leads to an increase in aromatase expression associated with an increase in CRTC nuclear localization, thereby providing a mechanism whereby PJS results in increased endogenous estrogens in affected males.


Assuntos
Aromatase/biossíntese , Ginecomastia/etiologia , Perda de Heterozigosidade , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/genética , Testículo/enzimologia , Quinases Proteína-Quinases Ativadas por AMP , Adolescente , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Pré-Escolar , Humanos , Masculino , Glândulas Mamárias Humanas/enzimologia , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Síndrome de Peutz-Jeghers/metabolismo , Síndrome de Peutz-Jeghers/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Células de Sertoli/enzimologia , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Testículo/metabolismo , Testículo/patologia , Fatores de Transcrição/metabolismo
20.
Spermatogenesis ; 1(3): 240-249, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22319672

RESUMO

Scientific discoveries over the past decade have shifted the stereotypical view of androgens as male hormones and estrogens as female hormones. It is now recognized that a delicate balance of both androgens and estrogens, a process controlled by aromatase, is fundamental for normal testicular development and fertility. While the site-specific actions of these two classes of steroids within the testis are becoming better documented, the role and regulation of estrogen biosynthesis by aromatase within the testis remains unclear. The majority of data comes from a wide range of animal species, particularly genetically modified mouse models; aromatase knockout (ArKO) and overexpressing (AROM(+)), with limited information on humans, however the existence of congenital aromatase mutations has provided some insight into its effects on individual parameters of the testis. This review dissects out the localization and activity of aromatase in the healthy and diseased testis, addresses the cellular insult to the testis that occurs in its absence and over abundance and proposes potential molecular mechanisms of aromatase regulation in the testis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA