Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 527-545, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27974247

RESUMO

Throughout spermatogenesis, two important processes occur at late stage VIII of the seminiferous epithelial cycle in the rat testis: preleptotene spermatocytes commence entry into the adluminal compartment and step 19 spermatids release from the seminiferous epithelium. Presently, it is not clear how these processes, which involve extensive restructuring of unique Sertoli-Sertoli and Sertoli-germ cell junctions, are mediated. We aimed to determine whether annexin A2 (ANXA2), a Ca2+-dependent and phospholipid-binding protein, participates in cell junction dynamics. To address this, in vitro and in vivo RNA interference studies were performed on prepubertal Sertoli cells and adult rat testes. The endpoints of Anxa2 knockdown were determined by immunoblotting, morphological analyses, fluorescent immunostaining, and barrier integrity assays. In the testis, ANXA2 localized to the Sertoli cell stalk, with specific staining at the blood-testis barrier and the concave (ventral) surface of elongated spermatids. ANXA2 also bound actin when testis lysates were used for immunoprecipitation. Anxa2 knockdown was found to disrupt the Sertoli cell/blood-testis barrier in vitro and in vivo. The disruption in barrier function was substantiated by changes in the localization of claudin-11, zona occludens-1, N-cadherin, and ß-catenin. Furthermore, Anxa2 knockdown resulted in spermiation defects caused by a dysfunction of tubulobulbar complexes, testis-specific actin-rich ultrastructures that internalize remnant cell junction components prior to spermiation. Additionally, there were changes in the localization of several tubulobulbar complex component proteins, including actin-related protein 3, cortactin, and dynamin I/II. Our results indicate that ANXA2 is critical for the integrity of the blood-testis barrier and the timely release of spermatids.


Assuntos
Anexina A2/genética , Barreira Hematotesticular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatogênese/genética , Proteína 3 Relacionada a Actina/genética , Proteína 3 Relacionada a Actina/metabolismo , Animais , Anexina A2/antagonistas & inibidores , Anexina A2/metabolismo , Barreira Hematotesticular/crescimento & desenvolvimento , Caderinas/genética , Caderinas/metabolismo , Claudinas/genética , Claudinas/metabolismo , Cortactina/genética , Cortactina/metabolismo , Dinamina I/genética , Dinamina I/metabolismo , Dinamina II/genética , Dinamina II/metabolismo , Junções Intercelulares/genética , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/citologia , Epitélio Seminífero/crescimento & desenvolvimento , Epitélio Seminífero/metabolismo , Células de Sertoli/citologia , Transdução de Sinais , Espermátides/crescimento & desenvolvimento , Espermátides/ultraestrutura , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Espermatócitos/ultraestrutura , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
2.
Results Probl Cell Differ ; 58: 225-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27300181

RESUMO

A healthy man typically produces between 50 × 10(6) and 200 × 10(6) spermatozoa per day by spermatogenesis; in the absence of Sertoli cells in the male gonad, this individual would be infertile. In the adult testis, Sertoli cells are sustentacular cells that support germ cell development by secreting proteins and other important biomolecules that are essential for germ cell survival and maturation, establishing the blood-testis barrier, and facilitating spermatozoa detachment at spermiation. In the fetal testis, on the other hand, pre-Sertoli cells form the testis cords, the future seminiferous tubules. However, the role of pre-Sertoli cells in this process is much less clear than the function of Sertoli cells in the adult testis. Within this framework, we provide an overview of the biology of the fetal, pubertal, and adult Sertoli cell, highlighting relevant cell biology studies that have expanded our understanding of mammalian spermatogenesis.


Assuntos
Células de Sertoli/citologia , Espermatogênese , Espermatozoides/citologia , Testículo/citologia , Animais , Barreira Hematotesticular/citologia , Barreira Hematotesticular/embriologia , Barreira Hematotesticular/crescimento & desenvolvimento , Diferenciação Celular , Humanos , Masculino , Túbulos Seminíferos/citologia , Túbulos Seminíferos/embriologia , Túbulos Seminíferos/crescimento & desenvolvimento , Testículo/embriologia , Testículo/crescimento & desenvolvimento
3.
Theriogenology ; 84(5): 763-72, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26074069

RESUMO

The formation of the blood-testis barrier (BTB) is defined as occurring with the first appearance of spermatocytes at around puberty and is vital for normal spermatogenesis. This barrier between two adjacent Sertoli cells (SCs) consists of a cell junctional protein complex, which includes tight junctions (TJs), adherens junctions, and gap junctions. In many mammalian species, BTB composition has already been investigated, whereas little is known about the equine BTB. In the present study, immunohistochemistry and qualitative Western Blot analysis were used to assess the expression and distribution patterns of the junctional proteins claudin-11 (TJ), zonula occludens-1 (TJ associated), N-cadherin (adherens junctions), and connexin 43 (gap junctions) in equine testes during tubular development and in testes of stallions exhibiting unilateral cryptorchidism. Therefore, testes of 21 warmblood stallions (aged 12 months-11 years) were obtained during routine surgical castration. In the normal adult equine testis, the junctional proteins are localized at the basolateral region of the seminiferous tubules forming a circumferential seal corresponding to the known BTB localization. N-cadherin is additionally expressed along the lateral SC surface. In immature seminiferous cords still lacking a lumen, a diffuse distribution pattern of the junctional proteins throughout the SC cytoplasm is visible. As lumen formation advances, the immunolocalization shifts progressively toward the basolateral SC membranes. Additionally, apoptotic germ cells were detected and quantified in prepubertal stallions using terminal deoxynucleotidyl transferase dUTP nick end labeling assay and correlated with junctional protein localization. In the retained testis of cryptorchid stallions, which exhibit an aberrant testicular morphology, a deviating expression of the junctional proteins is visible. The present data show for the first time that (1) the equine SC junctional complex contains claudin-11, zonula occludens-1, N-Cadherin, and connexin 43, as already described for men or mice, and that (2) different distribution patterns of these proteins exist during testicular development in the context of lumen formation (lumen scores: 1-7) and in retained testes of unilateral cryptorchid stallions.


Assuntos
Barreira Hematotesticular/crescimento & desenvolvimento , Cavalos/fisiologia , Animais , Barreira Hematotesticular/metabolismo , Western Blotting , Caderinas/metabolismo , Claudinas/metabolismo , Conexina 43/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Maturidade Sexual , Testículo/crescimento & desenvolvimento
4.
J Endocrinol ; 225(3): 125-34, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25876610

RESUMO

Development of the epididymis including blood-epididymal barrier formation is not required until sperm reach the epididymis peripuberally. Regulation of this development in the early postnatal period is largely unknown. The current objectives were to evaluate potential roles of endogenous estrogen and androgen signaling during early development of the corpus epididymidis and to determine the timing of formation of the blood-epididymal barrier in the pig. Effects of endogenous steroids were evaluated using littermates treated with vehicle, an aromatase inhibitor (letrozole) to reduce endogenous estrogens, an estrogen receptor antagonist (fulvestrant) or an androgen receptor antagonist (flutamide). Phosphorylated histone 3 immunohistochemistry was used to identify proliferating epithelial cells. Lanthanum nitrate and electron microscopy were used to analyze formation of the blood barrier in the corpus epididymidis. Reducing endogenous estrogens increased the number of proliferating corpus epithelial cells at 6 and 6.5 weeks of age compared with vehicle-treated boars (P<0.01 and P<0.001 respectively). Blocking androgen receptors did not alter proliferation rate at 6.5 weeks of age. Although barrier formation was similar between 6 and 6.5 weeks of age in vehicle-treated animals, intercellular barriers increased in letrozole-treated littermates at 6.5 weeks of age. Fulvestrant treatment, which should mimic aromatase inhibition for regulation through ESR1 and ESR2 signaling but potentially stimulate endogenous estrogen signaling through the G protein-coupled estrogen receptor (GPER), had the opposite effect on aromatase inhibition. These responses in conjunction with the presence of GPER in the corpus epididymidis suggest early corpus epididymal development is regulated partially by GPER.


Assuntos
Aromatase/metabolismo , Barreira Hematotesticular/crescimento & desenvolvimento , Epididimo/crescimento & desenvolvimento , Receptores Androgênicos/metabolismo , Desenvolvimento Sexual , Transdução de Sinais , Sus scrofa/crescimento & desenvolvimento , Antagonistas de Receptores de Andrógenos/farmacologia , Animais , Aromatase/química , Inibidores da Aromatase/farmacologia , Barreira Hematotesticular/efeitos dos fármacos , Barreira Hematotesticular/metabolismo , Barreira Hematotesticular/ultraestrutura , Epididimo/efeitos dos fármacos , Epididimo/metabolismo , Epididimo/ultraestrutura , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas do Receptor de Estrogênio/farmacologia , Flutamida/farmacologia , Fulvestranto , Letrozol , Masculino , Microscopia Eletrônica de Transmissão/veterinária , Nitrilas/farmacologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Receptores Androgênicos/química , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Desenvolvimento Sexual/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sus scrofa/fisiologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Triazóis/farmacologia
5.
PLoS One ; 9(8): e103809, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25118984

RESUMO

Nutrients and environmental chemicals, including endocrine disruptors, have been incriminated in the current increase in male reproductive dysfunction, but the underlying mechanisms remain unknown. The gastrointestinal tract represents the largest surface area exposed to our environment and thereby plays a key role in connection with exposure of internal organs to exogenous factors. In this context the gut microbiome (all bacteria and their metabolites) have been shown to be important contributors to body physiology including metabolism, cognitive functions and immunity. Pivotal to male reproduction is a proper development of the testis, including the formation of the blood-testis barrier (BTB) that encapsulates and protects germ cells from stress induced environmental cues, e.g. pathogenic organisms and xenobiotics. Here we used specific pathogen free (SPF) mice and germ-free (GF) mice to explore whether gut microbiota and/or their metabolites can influence testis development and regulation of BTB. Lumen formation in the seminiferous tubules, which coincides with the development of the BTB was delayed in the testes of GF mice at 16 days postpartum. In addition, perfusion experiments (Evans blue) demonstrated increased BTB permeability in these same mice. Reduced expressions of occludin, ZO-2 and E-cadherin in GF testis suggested that the microbiota modulated BTB permeability by regulation of cell-cell adhesion. Interestingly, exposure of GF mice to Clostridium Tyrobutyricum (CBUT), which secrete high levels of butyrate, restored the integrity of the BTB and normalized the levels of cell adhesion proteins. Moreover, the GF mice exhibited lower serum levels of gonadotropins (LH and FSH) than the SPF group. In addition, the intratesticular content of testosterone was lower in GF compared to SPF or CBUT animals. Thus, the gut microbiome can modulate the permeability of the BTB and might play a role in the regulation of endocrine functions of the testis.


Assuntos
Microbiota , Testículo/crescimento & desenvolvimento , Animais , Barreira Hematotesticular/crescimento & desenvolvimento , Adesão Celular , Fertilidade , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Permeabilidade , Organismos Livres de Patógenos Específicos
6.
Endocrinology ; 153(10): 5023-35, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22872576

RESUMO

The blood-testis barrier (BTB) is an important ultrastructure in the testis. A delay in its assembly during postnatal development leads to meiotic arrest. Also, a disruption of the BTB by toxicants in adult rats leads to a failure in spermatogonial differentiation. However, the regulation of BTB assembly remains unknown. Herein, filamin A, an actin filament cross-linker that is known to maintain and regulate cytoskeleton structure and function in other epithelia, was shown to be highly expressed during the assembly of Sertoli cell BTB in vitro and postnatal development of BTB in vivo, perhaps being used to maintain the actin filament network at the BTB. A knockdown of filamin A by RNA interference was found to partially perturb the Sertoli cell tight junction (TJ) permeability barrier both in vitro and in vivo. Interestingly, this down-regulating effect on the TJ barrier function after the knockdown of filamin A was associated with a mis-localization of both TJ and basal ectoplasmic specialization proteins. Filamin A knockdown also induced a disorganization of the actin filament network in Sertoli cells in vitro and in vivo. Collectively, these findings illustrate that filamin A regulates BTB assembly by recruiting these proteins to the microenvironment in the seminiferous epithelium to serve as the building blocks. In short, filamin A participates in BTB assembly by regulating protein recruitment during postnatal development in the rat testis.


Assuntos
Barreira Hematotesticular/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Junções Íntimas/metabolismo , Animais , Barreira Hematotesticular/crescimento & desenvolvimento , Células Cultivadas , Proteínas Contráteis/genética , Filaminas , Masculino , Proteínas dos Microfilamentos/genética , Permeabilidade , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Células de Sertoli/citologia , Testículo/citologia , Testículo/crescimento & desenvolvimento , Junções Íntimas/genética
7.
Am J Physiol Cell Physiol ; 293(1): C305-12, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17475664

RESUMO

A urea-selective urine-concentrating defect was found in transgenic mice deficient in urea transporter (UT)-B. To determine the role of facilitated urea transport in extrarenal organs expressing UT-B, we studied the kinetics of [(14)C]urea distribution in UT-B-null mice versus wild-type mice. After renal blood flow was disrupted, [(14)C]urea distribution was selectively reduced in testis in UT-B-null mice. Under basal conditions, total testis urea content was 335.4 +/- 43.8 microg in UT-B-null mice versus 196.3 +/- 18.2 microg in wild-type mice (P < 0.01). Testis weight in UT-B-null mice (6.6 +/- 0.8 mg/g body wt) was significantly greater than in wild-type mice (4.2 +/- 0.8 mg/g body wt). Elongated spermatids were observed earlier in UT-B-null mice compared with wild type mice on day 24 versus day 32, respectively. First breeding ages in UT-B knockout males (48 +/- 3 days) were also significantly earlier than that in wild-type males (56 +/- 2 days). In competing mating tests with wild-type males and UT-B-null males, all pups carried UT-B-targeted genes, which indicates that all pups were produced from breeding of UT-B-null males. Experiments of the expression of follicle-stimulating hormone receptor (FSHR) and androgen binding protein (ABP) indicated that the development of Sertoli cells was also earlier in UT-B-null mice than that in wild-type mice. These results suggest that UT-B plays an important role in eliminating urea produced by Sertoli cells and that UT-B deletion causes both urea accumulation in the testis and early maturation of the male reproductive system. The UT-B knockout mouse may be a useful experimental model to define the molecular mechanisms of early puberty.


Assuntos
Barreira Hematotesticular/metabolismo , Permeabilidade Capilar , Fertilidade , Proteínas de Membrana Transportadoras/metabolismo , Maturidade Sexual , Espermatogênese , Testículo/metabolismo , Ureia/metabolismo , Proteína de Ligação a Androgênios/genética , Proteína de Ligação a Androgênios/metabolismo , Animais , Barreira Hematotesticular/crescimento & desenvolvimento , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Tamanho do Órgão , RNA Mensageiro/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Comportamento Sexual Animal , Maturidade Sexual/genética , Espermátides/metabolismo , Espermátides/patologia , Espermatogênese/genética , Testículo/crescimento & desenvolvimento , Testículo/patologia , Fatores de Tempo , Ureia/sangue
8.
Anat Rec (Hoboken) ; 290(2): 206-14, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17441213

RESUMO

This research explores the initial assembly of the blood-testis barrier (BTB) during puberty, when a massive physiological apoptosis in the first spermatogenic wave takes place. Fragments of testis from 14- to 20-day-old rats were studied by conventional transmission electron microscopic techniques. Lanthanum hydroxide was used as an intercellular tracer. Light microscopy was used to confirm apoptotic death when paraffin-embedded sections were studied by TUNEL analysis. When the seminiferous cords reached the zygotene-pachytene spermatocyte level, they exhibited abundant apoptotic figures, whereas the remaining segments showed sporadic apoptosis. We found a BTB not yet assembled in the cords with zygotene-pachytene spermatocytes and abundant apoptosis. The observed apoptosis frequency diminished drastically when BTB was organized, as confirmed by the use of the tracer. Our conclusion is that the massive apoptosis found in the zygotene-pachytene spermatocytes between days 14 and 20 coincides with an open BTB. The absence of BTB could be one of the factors causing massive apoptosis of zygotene-pachytene spermatocytes, at least within the time span analyzed. The zygotene-pachytene spermatocytes are left exposed in an open environment instead of being isolated in the adluminal compartment to which they are destined.


Assuntos
Apoptose , Barreira Hematotesticular/ultraestrutura , Maturidade Sexual , Espermatócitos/ultraestrutura , Espermatogênese , Animais , Barreira Hematotesticular/crescimento & desenvolvimento , Masculino , Prófase Meiótica I , Microscopia Eletrônica de Transmissão , Estágio Paquíteno , Ratos , Ratos Wistar , Espermatócitos/crescimento & desenvolvimento , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...