Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Reprod Fertil Dev ; 29(3): 609-620, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26488911

RESUMEN

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.


Asunto(s)
Inhibinas/metabolismo , Tumor de Células de Sertoli/patología , Espermatogénesis/genética , Neoplasias Testiculares/patología , Activinas/sangre , Animales , Hormona Folículo Estimulante/sangre , Inhibinas/genética , Masculino , Ratones , Ratones Noqueados , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patología , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Testículo/metabolismo , Testículo/patología
2.
Reprod Fertil Dev ; 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26946412

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-25515817

RESUMEN

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.
Endocrinology ; 155(3): 1131-44, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424039

RESUMEN

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.


Asunto(s)
Barrera Hematotesticular/fisiología , Espermatogénesis , Animales , Claudinas/metabolismo , Hormona Folículo Estimulante/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Microscopía Fluorescente , Oligopéptidos/metabolismo , Tamaño de los Órganos , Permeabilidad , Ratas , Ratas Sprague-Dawley , Epitelio Seminífero/metabolismo , Espermátides/citología , Espermatocitos/citología , Testículo/metabolismo , Uniones Estrechas , Factores de Tiempo
5.
J Clin Endocrinol Metab ; 98(12): E1979-87, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24037887

RESUMEN

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.


Asunto(s)
Aromatasa/biosíntesis , Ginecomastia/etiología , Pérdida de Heterocigocidad , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinasas/genética , Testículo/enzimología , Quinasas de la Proteína-Quinasa Activada por el AMP , Adolescente , Núcleo Celular/metabolismo , Núcleo Celular/patología , Preescolar , Humanos , Masculino , Glándulas Mamarias Humanas/enzimología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Síndrome de Peutz-Jeghers/metabolismo , Síndrome de Peutz-Jeghers/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Células de Sertoli/enzimología , Células de Sertoli/metabolismo , Células de Sertoli/patología , Testículo/metabolismo , Testículo/patología , Factores de Transcripción/metabolismo
6.
Spermatogenesis ; 3(1): e24014, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23687617

RESUMEN

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.

7.
Biol Reprod ; 87(1): 13, 1-11, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492971

RESUMEN

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.


Asunto(s)
Células de Sertoli/citología , Animales , Comunicación Autocrina/fisiología , Proteínas de Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Humanos , Uniones Intercelulares/fisiología , Masculino , Modelos Biológicos , Comunicación Paracrina/fisiología , Pubertad/fisiología , Estaciones del Año , Células de Sertoli/fisiología , Maduración Sexual/fisiología , Transducción de Señal/fisiología , Espermatogénesis/fisiología
8.
Endocrinology ; 152(12): 4937-47, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22028441

RESUMEN

The conventional view is that estrogen confers female cardioprotection. Estrogen synthesis depends on androgen availability, with aromatase regulating conversion of testosterone to estradiol. Extragonadal aromatase expression mediates estrogen production in some tissues, but a role for local steroid conversion has not yet been demonstrated in the heart. This study's goal was to investigate how aromatase deficiency influences myocardial function and ischemic resilience. RT-PCR analysis of C57Bl/6 mouse hearts confirmed cardiac-specific aromatase expression in adult females. Functional performance of isolated hearts from female aromatase knockout (ArKO) and aromatase wild-type mice were compared. Left ventricular developed pressures were similar in aerobic perfusion, but the maximal rate of rise of ventricular pressure was modestly reduced in ArKO hearts (3725 ± 144 vs. 4272 ± 154 mm Hg/sec, P < 0.05). After 25 min of ischemia, the recovery of left ventricular developed pressure was substantially improved in ArKO (percentage of basal at 60 min of reperfusion, 62 ± 8 vs. 30 ± 6%; P < 0.05). Hypercontracture was attenuated (end diastolic pressure, 25 ± 5 vs. 51 ± 1 mm Hg; P < 0.05), and lactate dehydrogenase content of coronary effluent was reduced throughout reperfusion in ArKO hearts. This was associated with a hyperphosphorylation of phospholamban and a reduction in phosphorylated Akt. Immediately after reperfusion, ArKO hearts exhibited increased incidence of ventricular premature beats (194 ± 70 vs. 46 ± 6, P < 0.05). These observations indicate more robust functional recovery, reduced cellular injury, and modified cardiomyocyte Ca(2+) handling in aromatase-deficient hearts. Our findings indicate that androgen-to-estrogen conversion may be of pathophysiologic importance to the heart and challenge the notion that estrogen deficiency is deleterious. These studies suggest the possibility that aromatase suppression may offer inotropic benefit in the acute ischemia/reperfusion setting with appropriate arrhythmia management.


Asunto(s)
Aromatasa/deficiencia , Isquemia Miocárdica , Animales , Proteínas de Unión al Calcio/metabolismo , Cardiotónicos , Estradiol/deficiencia , Femenino , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Reperfusión Miocárdica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Presión Ventricular
9.
Biol Reprod ; 84(2): 379-91, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20926807

RESUMEN

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.


Asunto(s)
Activinas/metabolismo , Proliferación Celular , Feto/citología , Subunidades beta de Inhibinas/metabolismo , Células de Sertoli/citología , Espermatozoides/citología , Testículo/embriología , Receptores de Activinas/genética , Animales , Recuento de Células , Ciclo Celular/fisiología , Femenino , Fertilidad/fisiología , Desarrollo Fetal , Feto/anatomía & histología , Feto/metabolismo , Edad Gestacional , Subunidades beta de Inhibinas/genética , Masculino , Ratones , Tamaño de los Órganos , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo
10.
Spermatogenesis ; 1(3): 240-249, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22319672

RESUMEN

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.

11.
Endocrinology ; 151(6): 2911-22, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20357222

RESUMEN

Sertoli cell tight junctions (TJs) are an essential component of the blood-testis barrier required for spermatogenesis; however, the role of gonadotropins in their maintenance is unknown. This study aimed to investigate the effect of gonadotropin suppression and short-term replacement on TJ function and TJ protein (occludin and claudin-11) expression and localization, in an adult rat model in vivo. Rats (n = 10/group) received the GnRH antagonist, acyline, for 7 wk to suppress gonadotropins. Three groups then received for 7 d: 1) human recombinant FSH, 2) human chorionic gonadotropin (hCG) and rat FSH antibody (to study testicular androgen stimulation alone), and 3) hCG alone (to study testicular androgen and pituitary FSH production). TJ proteins were assessed by real-time PCR, Western blot analysis, and immunohistochemistry, whereas TJ function was assessed with a biotin permeation tracer. Acyline treatment significantly reduced testis weights, serum androgens, LH and FSH, and adluminal germ cells (pachytene spermatocyte, round and elongating spermatids). In contrast to controls, acyline induced seminiferous tubule permeability to biotin, loss of tubule lumens, and loss of occludin, but redistribution of claudin-11, immunostaining. Short-term hormone replacement stimulated significant recoveries in adluminal germ cell numbers. In hCG +/- FSH antibody-treated rats, occludin and claudin-11 protein relocalized at the TJ, but such relocalization was minimal with FSH alone. Tubule lumens also reappeared, but most tubules remained permeable to biotin tracer, despite the presence of occludin. It is concluded that gonadotropins maintain Sertoli cell TJs in the adult rat via a mechanism that includes the localization of occludin and claudin-11 at functional TJs.


Asunto(s)
Gonadotropinas/farmacología , Testículo/efectos de los fármacos , Testículo/metabolismo , Uniones Estrechas/metabolismo , Andrógenos/sangre , Animales , Western Blotting , Gonadotropina Coriónica/farmacología , Claudinas , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/farmacología , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Inmunohistoquímica , Hormona Luteinizante/sangre , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ocludina , Oligopéptidos/farmacología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Espermátides/efectos de los fármacos , Espermátides/metabolismo , Espermatocitos/efectos de los fármacos , Espermatocitos/metabolismo , Espermatogonias/efectos de los fármacos , Espermatogonias/metabolismo , Uniones Estrechas/efectos de los fármacos
12.
Mol Cell Endocrinol ; 323(2): 307-13, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20214950

RESUMEN

The enzyme aromatase converts androgens to estrogens, which have recently been postulated to be essential for testicular development and fertility. Understanding the mechanisms that regulate aromatase activity in the testis may therefore have implications for treatment of male infertility. Aromatase is encoded by the CYP19 gene, which uses multiple tissue-specific alternative promoters. In the testis, the proximal promoter PII drives aromatase expression. PII activity requires a nuclear receptor half-site, CAAGGTCA, to which two orphan receptors; SF-1 and LRH-1, have been shown to bind in vitro. The aim of this study was to investigate expression of aromatase and LRH-1 in the developing rat testis and define the ability of LRH-1 to induce aromatase expression in the testicular cells where both are expressed. We show that aromatase and LRH-1 are present throughout all stages of development of the rat testis, although the sites and levels of expression vary. The pattern of LRH-1 expression was broadly similar to that of aromatase. In adult animals higher levels of expression were observed in Leydig and germ cells. Over-expression of LRH-1 in primary rat Leydig and germ cells by adenoviral infection strongly increased endogenous aromatase mRNA levels, demonstrating the ability of LRH-1 to stimulate aromatase expression in vivo. We also observed binding of endogenous LRH-1 to the aromatase promoter II by chromatin immunoprecipitation. These data provide evidence that LRH-1 plays an important role in the regulation of testicular aromatase expression, and implicate LRH-1 as a regulator of rat spermatogenesis, in which estrogens are emerging as important mediators.


Asunto(s)
Aromatasa/metabolismo , Regulación Enzimológica de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/metabolismo , Testículo/enzimología , Testículo/crecimiento & desarrollo , Animales , Aromatasa/genética , Células Cultivadas , Células Germinativas/citología , Células Germinativas/metabolismo , Humanos , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/genética , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Testículo/citología
13.
J Endocrinol ; 205(2): 117-31, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20144980

RESUMEN

Over the past five decades, intense research using various animal models, innovative technologies notably genetically modified mice and wider use of stereological methods, unique agents to modulate hormones, genomic and proteomic techniques, have identified the cellular sites of spermatogenesis, that are regulated by FSH and testosterone. It has been established that testosterone is essential for spermatogenesis, and also FSH plays a valuable role. Therefore understanding the basic mechanisms by which hormones govern germ cell progression are important steps towards improved understating of fertility regulation in health diseases.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Infertilidad Masculina/metabolismo , Espermatozoides/crecimiento & desarrollo , Testosterona/metabolismo , Animales , Humanos , Masculino , Espermatogénesis , Espermatozoides/metabolismo
14.
Biol Reprod ; 82(5): 980-90, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20130270

RESUMEN

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.


Asunto(s)
Subunidades beta de Inhibinas/metabolismo , Células de Sertoli/metabolismo , Espermatogonias/metabolismo , Animales , Diferenciación Celular , Técnicas de Cocultivo , Células Germinativas/metabolismo , Subunidades beta de Inhibinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células de Sertoli/citología , Maduración del Esperma , Espermatogénesis/fisiología , Espermatogonias/citología , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo
15.
Int J Androl ; 32(5): 542-55, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18522674

RESUMEN

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.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Lipopolisacáridos/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Testículo/efectos de los fármacos , Animales , Secuencia de Bases , Celecoxib , Cartilla de ADN , Dinoprostona/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/patología , Testículo/fisiología
16.
Reproduction ; 135(6): 867-77, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18502899

RESUMEN

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.


Asunto(s)
Barrera Hematotesticular/metabolismo , Hormona Folículo Estimulante/fisiología , Proteínas de la Membrana/metabolismo , Phodopus/metabolismo , Testículo/metabolismo , Uniones Estrechas/metabolismo , Animales , Secuencia de Bases , Barrera Hematotesticular/efectos de los fármacos , Moléculas de Adhesión Celular/análisis , Moléculas de Adhesión Celular/genética , Claudina-3 , Claudinas , Cricetinae , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Hormona Folículo Estimulante/farmacología , Expresión Génica/efectos de los fármacos , Moléculas de Adhesión de Unión , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/genética , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Ocludina , Fosfoproteínas/análisis , Fosfoproteínas/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Estaciones del Año , Transcripción Genética , Proteína de la Zonula Occludens-1
17.
Med J Aust ; 188(9): 520-4, 2008 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-18459923

RESUMEN

OBJECTIVE: To report on the sentiments of the Australian health and medical research (HMR) workforce on issues related to employment and funding opportunities. DESIGN, SETTING AND PARTICIPANTS: In August 2006, the Australian Society for Medical Research (ASMR) invited all of its members to participate in an online survey. The survey took the form of a structured questionnaire that focused on career aspirations, career development and training opportunities, attitudes toward moving overseas to work, and employment conditions for medical researchers. MAIN OUTCOME MEASURES: Researchers' views on career opportunities, funding opportunities, salary and quality of the working environment; impact of these views on retaining a skilled medical research workforce in Australia. RESULTS: Of the 1258 ASMR members, 379 responded (30% response rate). Ninety-six per cent of respondents were currently based in Australia; 70% had a PhD or equivalent; and 58% were women. Most respondents worked at hospital research centres (37%), independent research institutes (28%) or university departments (24%). Sixty-nine per cent had funding from the National Health and Medical Research Council, with the remainder funded by other sources. Over the previous 5 years, 6% of respondents had left active research and 73% had considered leaving. Factors influencing decisions about whether to leave HMR included shortage of funding (91%), lack of career development opportunities (78%) and poor financial rewards (72%). Fifty-seven per cent of respondents were directly supported by grants or fellowships, with only 16% not reliant on grants for their continuing employment; 62% believed that funding had increased over the previous 5 years, yet only 30% perceived an increase in employment opportunities in HMR. Among the respondents, twice as many men as women held postgraduate qualifications and earned >or= dollars 100 000 a year. CONCLUSIONS: Employment insecurity and lack of funding are a cause of considerable anxiety among Australian health and medical researchers. This may have important implications for the recruitment and retention of researchers.


Asunto(s)
Actitud del Personal de Salud , Investigación Biomédica , Selección de Profesión , Financiación Gubernamental/economía , Empleos en Salud/estadística & datos numéricos , Sociedades Médicas/estadística & datos numéricos , Orientación Vocacional/métodos , Adulto , Australia , Investigación Biomédica/economía , Femenino , Humanos , Satisfacción en el Trabajo , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Sociedades Médicas/economía , Encuestas y Cuestionarios , Recursos Humanos
18.
J Endocrinol ; 197(1): 35-43, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18372230

RESUMEN

FSH is a key regulator of testis function, required for the establishment of full complements of Sertoli and germ cells during postnatal testis development and for the maintenance of spermatogenesis in the adult. FSH plays an important role in germ cell survival rather than proliferation, in the window between 14 and 18 days of testicular development, which coincides with the cessation of Sertoli cell proliferation and the onset of germ cell meiosis during the first wave of spermatogenesis. This study aimed to identify the pathway(s) of apoptosis regulated by changes in FSH levels in 14 - to 18-day-old rats, using a model of in vivo FSH suppression by passive immunoneutralization with a rat anti-FSH antibody. Apoptotic pathways were identified by immunohistochemistry using pathway-specific proteins as markers of the intrinsic (activated caspase 9) and extrinsic (activated caspase 8) pathways, followed by quantification of cell numbers using stereological techniques. In addition, RT-PCR was used to assess the expression of pathway-specific genes. We previously reported a 2.5-fold increase in spermatogonial apoptosis in these samples after 4 days of FSH suppression, and now show that this increase correlates with a 9.8-fold (P<0.001) increase in the frequency of caspase 9-positive spermatogonia in the absence of caspase 8 immunoreactivity. By contrast, spermatocytes exhibited both increased caspase 9 (7.5-fold; P<0.001) and caspase 8 (5.7 fold; P<0.001) immunoreactivities after 4 days of FSH suppression. No significant change in the transcription levels of candidate genes required for either pathway was detected. This study demonstrates that, in the seminiferous tubules, FSH suppression induces spermatogonial apoptosis predominantly via the intrinsic pathway, while spermatocyte apoptosis occurs via both the intrinsic and extrinsic pathways.


Asunto(s)
Apoptosis , Hormona Folículo Estimulante/fisiología , Espermatocitos/citología , Animales , Caspasa 8/genética , Caspasa 9/genética , Etiquetado Corte-Fin in Situ , Masculino , Ratas , Ratas Sprague-Dawley
19.
Hum Reprod ; 23(2): 403-11, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18199736

RESUMEN

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.


Asunto(s)
Gonadotropinas/fisiología , Espermatogonias/fisiología , Adulto , Apoptosis/fisiología , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Forma de la Célula , Supervivencia Celular/fisiología , Combinación de Medicamentos , Activación Enzimática , Gonadotropinas/antagonistas & inhibidores , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Acetato de Medroxiprogesterona/farmacología , Persona de Mediana Edad , Valores de Referencia , Espermátides/citología , Espermatogénesis/fisiología , Espermatogonias/citología , Espermatogonias/enzimología , Testosterona/farmacología
20.
Biol Reprod ; 78(4): 705-13, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18094353

RESUMEN

Follicle-stimulating hormone plays a key role in spermatogonial development in adult rats via poorly understood mechanisms. We aimed to identify the role of this hormone in the regulation of germ cell apoptosis and proliferation in adult rats by suppression of FSH action following passive immunoneutralization with a rat FSH antibody for 4 and 7 days. Apoptosis and proliferation were identified by TUNEL and proliferating cell nuclear antigen labeling methods, respectively. Intrinsic and extrinsic apoptotic pathways were identified by immunohistochemistry, stereological techniques, and RT-PCR by assessing pathway-specific proteins and genes. Following FSH suppression for 4 and 7 days, we have previously reported a 30% decrease in spermatogonial number, with increased apoptosis in a stage-specific manner. The present study also shows stage-specific increases in apoptosis with no changes in proliferation. This increase in apoptosis was attributable to an increase in spermatogonial apoptosis via the intrinsic rather than extrinsic pathway, as shown by increased activated caspase 9-positive spermatogonia. The concomitant suppression of FSH and LH/testosterone showed that testosterone alone or together with FSH was more important in spermatocyte and spermatid survival by regulating both apoptotic pathways. A reduction in the level of the intrinsic pathway transcript Bcl2l2 (apoptosis suppressor gene) following FSH suppression for 4 days shows that FSH regulates some components of the intrinsic pathway. This study reveals that FSH predominantly acts as a survival factor for spermatogonia by regulating the intrinsic pathway while having no affect on germ cell proliferation in rats in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Hormona Folículo Estimulante/fisiología , Espermatogonias/fisiología , Antagonistas de Andrógenos/farmacología , Animales , Anticuerpos/farmacología , Caspasa 9/metabolismo , División Celular , Activación Enzimática , Flutamida/farmacología , Hormona Folículo Estimulante/antagonistas & inhibidores , Hormona Folículo Estimulante/inmunología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/anatomía & histología , Testosterona/antagonistas & inhibidores , Testosterona/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...