Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
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.
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
5.
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
6.
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.

7.
Fertil Steril ; 100(3): 658-66, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23706332

RESUMO

OBJECTIVE: To assess the spatial organization of two proteins involved in the Sertoli cell junctional complex in men with primary seminiferous tubule failure. DESIGN: Retrospective study. SETTING: Medical research institute. PATIENT(S): Sixteen men total, six with meiotic arrest, seven with the Sertoli cell-only phenotype, and three with normal spermatogenesis. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Differences in claudin-11 and connexin-43 organization as detected using confocal microscopy. RESULT(S): In men with primary seminiferous tubule failure, four organizational patterns (I-IV) were recognized and quantified for claudin-11. Across these patterns, claudin-11 changed from a basal filamentous staining pattern to a punctate staining pattern with diffuse localization throughout the entire epithelium. Similar changes in staining patterns for connexin-43 were observed. Major differences were seen in the spatial organization of claudin-11 and connexin-43 in tubules from control men compared with tubules with primary seminiferous tubule failure, but we observed no differences in the spatial organization of these proteins in tubules from men with meiotic arrest and Sertoli cell-only phenotypes. CONCLUSION(S): The spatial organization of claudin-11 and connexin-43 is altered in men with primary seminiferous tubule failure. Disorganization of the proteins composing the Sertoli cell junctional complex may be involved in the spermatogenic impairment, possibly via loss of blood-testis barrier function.


Assuntos
Claudinas/metabolismo , Conexina 43/metabolismo , Infertilidade Masculina/metabolismo , Adulto , Estudos de Casos e Controles , Núcleo Celular/metabolismo , Humanos , Imuno-Histoquímica , Infertilidade Masculina/patologia , Masculino , Estudos Retrospectivos , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/patologia , Células de Sertoli , Espermatogênese/fisiologia , Distribuição Tecidual
8.
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
9.
Vet Surg ; 40(8): 952-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22091951

RESUMO

OBJECTIVE: To determine the incidence of medial meniscal tears in dogs with naturally occurring cranial cruciate ligament (CCL) disease treated with arthroscopy and tibial plateau leveling osteotomy (TPLO). STUDY DESIGN: Retrospective case series. SAMPLE POPULATION: Canine Stifles (n = 357) with naturally occurring CCL disease. METHODS: Medical records (November 2006-November 2009) were reviewed for all dogs with CCL disease treated with arthroscopic CCL debridement, meniscal probing, and TPLO. We investigated the significance of the preoperative variables; age, weight, tibial plateau angle (TPA), sex, and the preoperative condition of the CCL on the prevalence of concurrent meniscal tears (CMT) and incidence of late onset meniscal tears (LMT). RESULTS: Prevalence of CMT and incidence of LMT was 32.2% and 5.6%, respectively. A significant difference between age of dogs with and without CMT was identified. There was a significantly lower prevalence of CMT and incidence of LMT in dogs that had a partial CCL tear compared with those that had a complete CCL tear. All dogs with LMT treated by partial meniscectomy (PMM) returned to peak postoperative limb function after (PMM) based on client-assessed outcomes. CONCLUSIONS: This study demonstrates the importance of LMT as a complication, the importance of comprehensive meniscal assessment, and may add to the evidence against routinely performing meniscal release in TPLO. The preoperative condition of the CCL should be considered before operating on grossly normal menisci.


Assuntos
Artroscopia/veterinária , Doenças do Cão/cirurgia , Artropatias/veterinária , Meniscos Tibiais/patologia , Osteotomia/veterinária , Complicações Pós-Operatórias/veterinária , Tíbia/cirurgia , Animais , Cães , Feminino , Incidência , Artropatias/cirurgia , Ligamentos Articulares/patologia , Masculino , Osteotomia/métodos , Complicações Pós-Operatórias/epidemiologia , Estudos Retrospectivos , Joelho de Quadrúpedes/patologia , Joelho de Quadrúpedes/cirurgia , Resultado do Tratamento
10.
Endocrinology ; 152(12): 4937-47, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22028441

RESUMO

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.


Assuntos
Aromatase/deficiência , Isquemia Miocárdica , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cardiotônicos , Estradiol/deficiência , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Reperfusão Miocárdica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pressão Ventricular
11.
Med Educ ; 45(2): 200-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21208265

RESUMO

OBJECTIVES: this study aims to project attrition from the Australian health and medical research workforce for those aged > 40 years in 2009, through to 2019, and to draw conclusions about the future of this workforce and the international implications of ageing workforce populations. METHODS: the study uses recently collected unpublished demographic data on the 2009 health and medical research workforce drawn from an Australian Society for Medical Research survey of health and medical research organisations. RESULTS: about 6250 members of the health and medical research workforce aged > 40 years in 2009 are expected to leave the workforce during 2009-2019; the bulk of these will be aged 50-69 years. It is estimated that 35% of women and 49% of men aged 40-49 years in 2009 will retire by the age of 50-59 years, and 85% of women and 70% of men aged 50-59 years in 2009 are also projected to retire over the next 10 years. Of the 6250 members who are expected to leave the workforce by 2019, about 4000 hold a PhD. As a result of population growth, a further 1700 persons with a PhD will be required if Australia is to maintain its current ratio of PhD-qualified persons in the health and medical research workforce: working population to 2019, at a cost of about AU$240 million. CONCLUSIONS: there is a need to plan for the replacement of the retiring generation of the health and medical research workforce and for the growth required to match that of the working population. If Australia is to fulfil its ambition for a highly educated, optimally skilled and highly trained health and medical research sector, it must heighten its focus on the higher education of young medical researchers. As population ageing is an emerging phenomenon worldwide, all first world nations are likely to face the challenges involved in replacing a rapidly retiring generation of the health and medical research workforce.


Assuntos
Pesquisa Biomédica/tendências , Educação de Pós-Graduação em Medicina/organização & administração , Adulto , Distribuição por Idade , Idoso , Austrália , Pesquisa Biomédica/educação , Pesquisa Biomédica/estatística & dados numéricos , Escolaridade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Avaliação das Necessidades , Aposentadoria/estatística & dados numéricos , Aposentadoria/tendências , Recursos Humanos , Adulto Jovem
12.
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
13.
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.

14.
Int J Radiat Oncol Biol Phys ; 77(3): 886-94, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20510199

RESUMO

PURPOSE: High-dose synchrotron microbeam radiation therapy (MRT) can be effective at destroying tumors in animal models while causing very little damage to normal tissues. The aim of this study was to investigate the cellular processes behind this observation of potential clinical importance. METHODS AND MATERIALS: MRT was performed using a lattice of 25 mum-wide, planar, polychromatic, kilovoltage X-ray microbeams, with 200-microm peak separation. Inoculated EMT-6.5 tumor and normal mouse skin tissues were harvested at defined intervals post-MRT. Immunohistochemical detection of gamma-H2AX allowed precise localization of irradiated cells, which were also assessed for proliferation and apoptosis. RESULTS: MRT significantly reduced tumor cell proliferation by 24 h post-irradiation (p = 0.002). An unexpected finding was that within 24 h of MRT, peak and valley irradiated zones were indistinguishable in tumors because of extensive cell migration between the zones. This was not seen in MRT-treated normal skin, which appeared to undergo a coordinated repair response. MRT elicited an increase in median survival times of EMT-6.5 and 67NR tumor-inoculated mice similar to that achieved with conventional radiotherapy, while causing markedly less normal tissue damage. CONCLUSIONS: This study provides evidence of a differential response at a cellular level between normal and tumor tissues after synchrotron MRT.


Assuntos
Histonas/análise , Neoplasias Mamárias Experimentais/radioterapia , Lesões Experimentais por Radiação/prevenção & controle , Tolerância a Radiação/fisiologia , Pele/efeitos da radiação , Síncrotrons , Animais , Apoptose/fisiologia , Biomarcadores/análise , Movimento Celular/fisiologia , Movimento Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Quebras de DNA de Cadeia Dupla , Reparo do DNA/fisiologia , Feminino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Lesões Experimentais por Radiação/metabolismo , Tolerância a Radiação/genética , Distribuição Aleatória , Pele/citologia , Fatores de Tempo
15.
Endocrinology ; 151(6): 2911-22, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20357222

RESUMO

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.


Assuntos
Gonadotropinas/farmacologia , Testículo/efeitos dos fármacos , Testículo/metabolismo , Junções Íntimas/metabolismo , Androgênios/sangue , Animais , Western Blotting , Gonadotropina Coriônica/farmacologia , Claudinas , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Imuno-Histoquímica , Hormônio Luteinizante/sangue , Masculino , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Ocludina , Oligopeptídeos/farmacologia , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Espermátides/efeitos dos fármacos , Espermátides/metabolismo , Espermatócitos/efeitos dos fármacos , Espermatócitos/metabolismo , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Junções Íntimas/efeitos dos fármacos
16.
Mol Cell Endocrinol ; 323(2): 307-13, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20214950

RESUMO

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.


Assuntos
Aromatase/metabolismo , Regulação Enzimológica da Expressão Gênica , Receptores Citoplasmáticos e Nucleares/metabolismo , Testículo/enzimologia , Testículo/crescimento & desenvolvimento , Animais , Aromatase/genética , Células Cultivadas , Células Germinativas/citologia , Células Germinativas/metabolismo , Humanos , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Masculino , Camundongos , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/genética , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Testículo/citologia
17.
J Endocrinol ; 205(2): 117-31, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20144980

RESUMO

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.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Infertilidade Masculina/metabolismo , Espermatozoides/crescimento & desenvolvimento , Testosterona/metabolismo , Animais , Humanos , Masculino , Espermatogênese , Espermatozoides/metabolismo
18.
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
19.
Am J Pathol ; 174(1): 184-95, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095948

RESUMO

Activin A is a potent growth and differentiation factor whose synthesis and bioactivity are tightly regulated. Both follistatin binding and inhibin subunit heterodimerization block access to the activin receptor and/or receptor activation. We postulated that the activin-beta(C) subunit provides another mechanism regulating activin bioactivity. To test our hypothesis, we examined the biological effects of activin C and produced mice that overexpress activin-beta(C). Activin C reduced activin A bioactivity in vitro; in LNCaP cells, activin C abrogated both activin A-induced Smad signaling and growth inhibition, and in LbetaT2 cells, activin C antagonized activin A-mediated activity of an follicle-stimulating hormone-beta promoter. Transgenic mice that overexpress activin-betaC exhibited disease in testis, liver, and prostate. Male infertility was caused by both reduced sperm production and impaired sperm motility. The livers of the transgenic mice were enlarged because of an imbalance between hepatocyte proliferation and apoptosis. Transgenic prostates showed evidence of hypertrophy and epithelial cell hyperplasia. Additionally, there was decreased evidence of nuclear Smad-2 localization in the testis, liver, and prostate, indicating that overexpression of activin-beta(C) antagonized Smad signaling in vivo. Underlying the significance of these findings, human testis, liver, and prostate cancers expressed increased activin-betaC immunoreactivity. This study provides evidence that activin-beta(C) is an antagonist of activin A and supplies an impetus to examine its role in development and disease.


Assuntos
Subunidades beta de Inibinas/metabolismo , Animais , Western Blotting , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Infertilidade Masculina , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad2/metabolismo , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Análise Serial de Tecidos , Transfecção
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...