Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 309
Filtrar
1.
J Anim Sci ; 1012023 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37282598

RESUMO

Minks are seasonal breeders whose seminiferous epithelium undergoes regression through massive germ cell death, leaving only Sertoli cells and spermatogonial cells in the tubules. However, the molecular mechanisms that control this biological process remain largely unknown. This study describes a transcriptomic analysis of mink testes at various reproductive stages (active, regressing, and inactive). A comparison of seminiferous epithelium at different stages of reproduction shows that cell adhesion is altered during regression. In addition, genes and proteins involved in forming the blood-testis barrier (BTB) were examined in sexually active and inactive minks. The seminiferous epithelium in the testes of sexually inactive minks expressed occludin, but this expression was not discernibly observed in the testes of sexually active minks. There was no discernible expression of CX43 in the seminiferous epithelium in the testes of sexually inactive minks, but CX43 was expressed in the testes of sexually active minks. During the regression process, we observed a remarkable increase in the expression levels of Claudin-11, which is associated with Sertoli-germ cell junctions. In conclusion, these findings suggest a loss of Sertoli-germ cell adhesion, which may regulate postmeiotic cell shedding during testicular regression in mink.


Here, we report for the first time the molecular mechanisms of testicular regression in mink. Our results, together with studies on other animals' characteristic reproductive features, identify a cluster of events crucial to the seminiferous epithelium regression process in mammalian seasonal breeders and highlight perspectives unique to the mink.


Assuntos
Vison , Epitélio Seminífero , Masculino , Animais , Epitélio Seminífero/metabolismo , Adesão Celular , Estações do Ano , Conexina 43/metabolismo , Testículo/metabolismo , Células de Sertoli/metabolismo
2.
Mol Cell Endocrinol ; 571: 111936, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37119967

RESUMO

Four-jointed box kinase 1 (Fjx1) is a planar cell polarity (PCP) protein and a member of the Fat (FAT atypical cadherin 1)/Dchs (Dachsous cadherin-related protein)/Fjx1 PCP complex. Fjx1 is also a non-receptor Ser/Thr protein kinase capable of phosphorylating Fat1 at is extracellular cadherin domains when it is being transported across the Golgi system. As such, Fjx1 is a Golgi-based regulator of Fat1 function by determining its extracellular deposition. Herein, Fjx1 was found to localize across the Sertoli cell cytoplasm, partially co-localized with the microtubules (MTs) across the seminiferous epithelium. It was most notable at the apical ES (ectoplasmic specialization) and basal ES, displaying distinctive stage-specific expression. The apical ES and basal ES are the corresponding testis-specific cell adhesion ultrastructures at the Sertoli-elongated spermatid interface and the Sertoli cell-cell interface, respectively, consistent with the role of Fjx1 as a Golgi-associated Ser/Thr kinase that modulates the Fat (and/or Dchs) integral membrane proteins. Its knockdown (KD) by RNAi using specific Fjx1 siRNA duplexes versus non-targeting negative control siRNA duplexes was found to perturb the Sertoli cell tight junction function, as well as perturbing the function and organization of MT and actin. While Fjx1 KD did not affect the steady-state levels of almost two dozens of BTB-associated Sertoli cell proteins, including structural and regulatory proteins, its KD was found to down-regulate Fat1 (but not Fat2, 3, and 4) and to up-regulate Dchs1 (but not Dchs2) expression. Based on results of biochemical analysis, Fjx1 KD was found to be capable of abolishing phosphorylation of its putative substrate Fat1 at its Ser/Thr sites, but not at its Tyr site, illustrating an intimate functional relationship of Fjx1 and Fat1 in Sertoli cells.


Assuntos
Células de Sertoli , Espermatogênese , Ratos , Animais , Masculino , Células de Sertoli/metabolismo , Espermatogênese/genética , Polaridade Celular , Ratos Sprague-Dawley , Testículo/metabolismo , Epitélio Seminífero/metabolismo , Caderinas/metabolismo , RNA Interferente Pequeno/metabolismo , Barreira Hematotesticular/metabolismo
3.
Andrology ; 11(5): 904-910, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36793255

RESUMO

Precise spatiotemporal expression of cohorts of differentiation markers unique to spermatogonia, spermatocytes, and round spermatids punctuates spermatogenesis and ensures its completion. For example, genes coding for the synaptonemal complex or the acrosome or flagellum are expressed sequentially in a developmental stage- and germ cell-specific manner. But the transcriptional mechanisms governing the spatiotemporal order of gene expression within the seminiferous epithelium are poorly understood. Using the round spermatid-specific Acrv1 gene, which codes for the acrosomal protein SP-10 as a model, we learned that (1) the proximal promoter itself contains all the necessary cis-regulatory sequences, (2) an insulator prevents somatic cell expression of the testis-specific gene, (3) RNA II polymerase is loaded on the Acrv1 promoter but paused in spermatocytes, thus ensuring precise transcriptional elongation in round spermatids, and that (4) a transcriptional repressor binding protein of 43 kilodaltons (TDP-43) plays a role in maintaining the paused state in spermatocytes. Although the Acrv1 enhancer element has been narrowed down to 50 bp and its binding to a 47 kDa testis-abundant nuclear protein shown, the identity of the putative transcription factor responsible for activation of round spermatid-specific transcription remains elusive. Human male infertility is idiopathic with limited treatment options. Understanding transcriptional regulation of spermatogenesis has the potential to lead to future therapies for male infertility.


Assuntos
Infertilidade Masculina , Epitélio Seminífero , Camundongos , Masculino , Humanos , Animais , Epitélio Seminífero/metabolismo , Proteínas de Membrana/genética , Espermátides/metabolismo , Regulação da Expressão Gênica , Espermatogênese/genética , Testículo/metabolismo , Infertilidade Masculina/genética , Expressão Gênica
4.
J Mol Endocrinol ; 69(4): T51-T57, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35670629

RESUMO

Spermatogenesis is a highly organized and regulated process that requires the constant production of millions of gametes over the reproductive lifetime of the mammalian male. This is possible because of an active stem cell pool and an ordered entry into the germ cell developmental sequence. The ordered entry is a result of the synthesis and action of retinoic acid allowing for the onset of spermatogonial differentiation and an irreversible commitment to spermatogenesis. The periodic appearance and actions of retinoic acid along the seminiferous tubules is a result of the interactions between germ cells and Sertoli cells that result in the generation and maintenance of the cycle of the seminiferous epithelium and is the subject of this review.


Assuntos
Espermatogênese , Tretinoína , Animais , Masculino , Mamíferos/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Testículo/metabolismo , Tretinoína/metabolismo , Tretinoína/farmacologia
5.
Histol Histopathol ; 37(7): 621-636, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35388905

RESUMO

The PI3K/AKT signaling pathway is one of the most crucial regulatory mechanisms in animal cells, which can mainly regulate proliferation, survival and anti-apoptosis in cell lines. In the seminiferous epithelium, most studies were concentrated on the role of PI3K/AKT signaling in immature Sertoli cells (SCs) and spermatogonia stem cells (SSCs). PI3K/AKT signaling can facilitate the proliferation and anti-apoptosis of immature Sertoli cells and spermatogenic cells. Besides, in mature Sertoli cells, this pathway can disintegrate the structure of the blood-testis barrier (BTB) via regulatory protein synthesis and the cytoskeleton of Sertoli cells. All of these effects can directly and indirectly maintain and promote spermatogenesis in male testis.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Células de Sertoli , Animais , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Transdução de Sinais , Espermatogênese/fisiologia , Testículo
6.
Reprod Fertil Dev ; 34(7): 560-575, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35143740

RESUMO

CONTEXT: Juxtacrine (contact-dependent) communication between the cells of seminiferous epithelium mediated by Notch signalling is of importance for the proper course of spermatogenesis in mammals. AIMS: The present study was designed to evaluate the role of follicle-stimulating hormone (FSH) in the regulation of Notch signalling in rodent seminiferous epithelium. METHODS: We explored the effects (1) of pharmacological inhibition of the hypothalamus-pituitary-gonadal (HPG) axis and FSH replacement in pubertal rats, and (2) of photoinhibition of HPG axis followed by FSH substitution in seasonally breeding rodents, bank voles, on Notch pathway activity. Experiments on isolated rat Sertoli cells exposed to FSH were also performed. Gene and protein expressions of Notch pathway components were analysed using RT-qPCR, western blot and immunohistochemistry/immunofluorescence. KEY RESULTS: Distribution patterns of Notch pathway proteins in bank vole and rat seminiferous epithelium were comparable; however, levels of activated Notch1 and Notch3, hairy/enhancer of split 1 (HES1) and hairy/enhancer of split-related with YRPW motif 1 (HEY1) in bank voles were dependent on the length of the photoperiod. In response to FSH similar changes in these proteins were found in both species, indicating that FSH is a negative regulator of Notch pathway activity in seminiferous epithelium. CONCLUSIONS: Our results support a common mechanism of FSH action on Notch pathway during onset and recrudescence of spermatogenesis in rodents. IMPLICATIONS: Interaction between FSH signalling and Notch pathway in Sertoli cells may be involved in spermatogenic activity changes of the testes occurring during puberty or photoperiod shift in continuously and seasonally breeding rodents, respectively.


Assuntos
Hormônio Foliculoestimulante , Epitélio Seminífero , Animais , Hormônio Foliculoestimulante/farmacologia , Masculino , Ratos , Receptores Notch/metabolismo , Roedores/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatogênese , Testículo/metabolismo
7.
Andrology ; 10(2): 377-391, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34535976

RESUMO

BACKGROUNDS: Sterility induced by anti-cancer treatments has caused significant concern, yet the mechanism and treatment exploration are little for male infertility after cancer therapy. Busulfan, the antineoplastic that was widely applied before bone marrow transplantation, was known to induce male reproductive disorder. OBJECTIVES: To investigate the effect of busulfan on blood-testis barrier function in adult rats and determine whether noncollagenous 1 domain peptide, the biologically active fragment proteolyzed from the collagen α3 chain (IV) by matrix metalloproteinase 9, was involved during this process. MATERIALS AND METHODS: Adult male rats were treated with one-dose or double-dose of busulfan (10 mg/kg) before euthanized at day 35. Blood-testis barrier integrity assay, HE staining, immunofluorescence, and Western blot were used to validate the effect of busulfan on blood-testis barrier permeability and spermatogenesis. JNJ0966 was applied to specifically inhibit the matrix metalloproteinase 9 activity. The polymerization activity of F-actin/G-actin and microtubule/tubulin in the testis were assessed by using commercial kits. RESULTS: A noteworthy blood-testis barrier injury and significant up-regulation of matrix metalloproteinase 9 activity and noncollagenous 1 level after a single-dose busulfan (10 mg/kg) treatment in adult rat testis were revealed. The application of JNJ0966 was found to decrease noncollagenous 1 level and rescue the busulfan-induced blood-testis barrier injury including the mis-localization of junction proteins across the seminiferous epithelium, by recovering the organization and polymerization of both F-actin and microtubule. The busulfan-induced spermatogenesis impairment was also improved by JNJ0966. CONCLUSION: These findings thus demonstrate that the elevation in matrix metalloproteinase 9 and noncollagenous 1 might participate in busulfan-induced blood-testis barrier disruption in adult male rats. As such, busulfan-induced male infertility could possibly be managed through interventions on noncollagenous 1 production.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Barreira Hematotesticular/efeitos dos fármacos , Bussulfano/efeitos adversos , Infertilidade Masculina/induzido quimicamente , Espermatogênese/efeitos dos fármacos , Animais , Autoantígenos/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Ratos , Epitélio Seminífero/metabolismo
8.
J Biol Chem ; 297(5): 101231, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34599968

RESUMO

Meiotic arrest is a common cause of human male infertility, but the causes of this arrest are poorly understood. Transactive response DNA-binding protein of 43 kDa (TDP-43) is highly expressed in spermatocytes in the preleptotene and pachytene stages of meiosis. TDP-43 is linked to several human neurodegenerative disorders wherein its nuclear clearance accompanied by cytoplasmic aggregates underlies neurodegeneration. Exploring the functional requirement for TDP-43 for spermatogenesis for the first time, we show here that conditional KO (cKO) of the Tardbp gene (encoding TDP-43) in male germ cells of mice leads to reduced testis size, depletion of germ cells, vacuole formation within the seminiferous epithelium, and reduced sperm production. Fertility trials also indicated severe subfertility. Spermatocytes of cKO mice showed failure to complete prophase I of meiosis with arrest at the midpachytene stage. Staining of synaptonemal complex protein 3 and γH2AX, markers of the meiotic synaptonemal complex and DNA damage, respectively, and super illumination microscopy revealed nonhomologous pairing and synapsis defects. Quantitative RT-PCR showed reduction in the expression of genes critical for prophase I of meiosis, including Spo11 (initiator of meiotic double-stranded breaks), Rec8 (meiotic recombination protein), and Rad21L (RAD21-like, cohesin complex component), as well as those involved in the retinoic acid pathway critical for entry into meiosis. RNA-Seq showed 1036 upregulated and 1638 downregulated genes (false discovery rate <0.05) in the Tardbp cKO testis, impacting meiosis pathways. Our work reveals a crucial role for TDP-43 in male meiosis and suggests that some forms of meiotic arrest seen in infertile men may result from the loss of function of TDP-43.


Assuntos
Proteínas de Ligação a DNA/deficiência , Regulação da Expressão Gênica , Infertilidade Masculina/metabolismo , Prófase Meiótica I , Epitélio Seminífero/metabolismo , Espermatócitos/metabolismo , Espermatogênese , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout
9.
Reprod Biol ; 21(4): 100562, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34555686

RESUMO

The structural integrity of the germ cells in the seminiferous epithelium and the correct process of spermatogenesis are made possible by proteins that participate in the formation of different types of junctions. This study was performed on samples of the testes of 4 groups (2 experimental and 2 corresponding control) of male Wistar rats. In the first experimental group, the adult rats received letrozole - a nonsteroidal inhibitor of cytochrome P450 aromatase (P450arom). The second experimental group was exposed to soya isoflavones during the prenatal period, lactation, and up to sexual maturity. The aim of this study was to examine the immunoexpression of ß-catenin, N-cadherin, occludin, connexin43, annexin V, and advanced glycation end products (AGE) in the seminiferous epithelium of rat testes with chronic estrogen deficiency and of rats exposed to soya isoflavones. Series of sections of the testes were stained using PAS and silver impregnation. Moreover, immunohistochemistry tests were performed. A semi-quantitative determination of protein immunoexpression was performed using Image J. The number of annexin V positive Sertoli cells per tubule were counted manually. Comparisons between the experimental and corresponding control groups were performed using a non-parametric Mann-Whitney U test. The most common alterations were prematurely sloughed germ cells in the lumen of the seminiferous tubules and invaginations of the seminiferous tubules. We observed a lower number of annexin V positive Sertoli cells and a lower expression of N-cadherin and occludin in the seminiferous epithelium of both groups of rats with hormonal imbalances. Moreover, a higher expression of AGE, a lower expression of connexin 43 and a lower amount of reticular fibers in the basal lamina of seminiferous tubules was present in rats treated with letrozole and a higher expression of ß-catenin was found in rats exposed to soya isoflavones. The hormonal imbalance between androgens and estrogens resulted in a decreased number of annexin V positive Sertoli cells. This may be associated with a failed clearance of apoptotic germ cells that leads to disturbances in the blood-testis-barrier (BTB) by affecting the expression of junctional proteins in the seminiferous epithelium. Moreover, a decreased level of estrogens was also associated with an increased expression of AGEs and with a changed composition of basal lamina in the seminiferous tubules of rats. These changes could lead to germ cell sloughing and invaginations of the seminiferous tubules.


Assuntos
Estrogênios/deficiência , Junções Intercelulares/metabolismo , Isoflavonas/farmacologia , Proteínas de Membrana/metabolismo , Epitélio Seminífero/metabolismo , Animais , Barreira Hematotesticular/efeitos dos fármacos , Feminino , Produtos Finais de Glicação Avançada/metabolismo , Letrozol , Masculino , Exposição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos Wistar , Epitélio Seminífero/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos
10.
Endocrinology ; 162(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34338758

RESUMO

The blood-testis barrier (BTB) and apical ectoplasmic specialization (ES), which are synchronized through the crosstalk of Sertoli cells and Sertoli germ cells, are required for spermatogenesis and sperm release. Here, we show that Wnt5a, a noncanonical Wnt signaling pathway ligand, is predominately expressed in both the BTB and apical ES and has a specific expression pattern during the seminiferous epithelium cycle. We employed siRNA to knockdown Wnt5a expression in testis and Sertoli cells, and then identified elongated spermatids that lost their polarity and were embedded in the seminiferous epithelium. Moreover, phagosomes were found near the tubule lumen. These defects were due to BTB and apical ES disruption. We also verified that the expression level and/or location of BTB-associated proteins, actin binding proteins (ABPs), and F-actin was changed after Wnt5a knockdown in vivo and in vitro. Additionally, we demonstrated that Wnt5a regulated actin dynamics through Ror2-mediated mTORC1 and mTORC2. This study clarified the molecular mechanism of Wnt5a in Sertoli cell junctions through the planar cell polarity (PCP) signaling pathway. Our findings could provide an experimental basis for the clinical diagnosis and treatment of male infertility caused by Sertoli cell junction impairment.


Assuntos
Regulação da Expressão Gênica , Alvo Mecanístico do Complexo 1 de Rapamicina/biossíntese , Alvo Mecanístico do Complexo 2 de Rapamicina/biossíntese , Células de Sertoli/metabolismo , Proteína Wnt-5a/biossíntese , Citoesqueleto de Actina/metabolismo , Animais , Barreira Hematotesticular , Perfilação da Expressão Gênica , Células Germinativas/citologia , Ligantes , Masculino , Proteínas dos Microfilamentos/metabolismo , Fagossomos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Epitélio Seminífero/metabolismo , Transdução de Sinais , Espermátides/metabolismo , Espermatogênese/genética , Testículo/metabolismo , Proteína Wnt-5a/metabolismo
11.
PLoS One ; 16(5): e0251256, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34014973

RESUMO

Although rodents represent approximately 40% of all living mammalian species, our knowledge regarding their reproductive biology is still scarce. Due to their high vulnerability to environmental changes, wild rodents have become beneficial models for ecological studies. Thus, we aimed to comparatively investigate key functional testis parameters in four sexually mature wild rodent species (A. cursor, A. montensis, N. lasiurus, and O. nigripes). These species belong to the Cricetidae family, which is the most diverse family of rodents in South America, with a total of ~120 species in Brazil. The results found for the gonadosomatic index and the sickled sperm head shape observed strongly suggest that the species here evaluated are promiscuous, prolific, and short-lived. The duration of spermatogenesis was relatively short and varied from ~35-40 days. Both the percentage of seminiferous tubules (ST) in the testis parenchyma (~95-97%) and the number of Sertoli cells (SC) (~48-70 million) per testis gram were very high, whereas a fairly good SC efficiency (~8-13 round spermatids per SC) was observed. In comparison to other mammalian species studied, particularly the rodents of the suborder Myomorpha (i.e. hamsters, rats and mice), the rodents herein investigated exhibited very high (~62-80 million) daily sperm production per testis gram. This impressive spermatogenic efficiency resulted mainly from the short duration of spermatogenesis and quite high values found for the ST percentage in the testis and the SC number per testis gram. We expect that the knowledge here obtained will help conservation programs and the proper management of wildlife.


Assuntos
Arvicolinae/metabolismo , Espermatogênese/fisiologia , Testículo/citologia , Animais , Arvicolinae/fisiologia , Brasil , Células Intersticiais do Testículo/metabolismo , Masculino , Epitélio Seminífero/metabolismo , Túbulos Seminíferos/metabolismo , Células de Sertoli/metabolismo , Espermátides/metabolismo , Espermatozoides/metabolismo
12.
Crit Rev Biochem Mol Biol ; 56(3): 236-254, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33761828

RESUMO

It is almost five decades since the discovery of the hypothalamic-pituitary-testicular axis. This refers to the hormonal axis that connects the hypothalamus, pituitary gland and testes, which in turn, regulates the production of spermatozoa through spermatogenesis in the seminiferous tubules, and testosterone through steroidogenesis by Leydig cells in the interstitium, of the testes. Emerging evidence has demonstrated the presence of a regulatory network across the seminiferous epithelium utilizing bioactive molecules produced locally at specific domains of the epithelium. Studies have shown that biologically active fragments are produced from structural laminin and collagen chains in the basement membrane. Additionally, bioactive peptides are also produced locally in non-basement membrane laminin chains at the Sertoli-spermatid interface known as apical ectoplasmic specialization (apical ES, a testis-specific actin-based anchoring junction type). These bioactive peptides are derived from structural laminins and/or collagens at the corresponding sites through proteolytic cleavage by matrix metalloproteinases (MMPs). They in turn serve as autocrine and/or paracrine factors to modulate and coordinate cellular events across the epithelium by linking the apical and basal compartments, the apical and basal ES, the blood-testis barrier (BTB), and the basement membrane of the tunica propria. The cellular events supported by these bioactive peptides/fragments include the release of spermatozoa at spermiation, remodeling of the immunological barrier to facilitate the transport of preleptotene spermatocytes across the BTB, and the transport of haploid spermatids across the epithelium to support spermiogenesis. In this review, we critically evaluate these findings. Our goal is to identify research areas that deserve attentions in future years. The proposed research also provides the much needed understanding on the biology of spermatogenesis supported by a local network of regulatory biomolecules.


Assuntos
Barreira Hematotesticular/metabolismo , Colágeno/metabolismo , Epitélio Seminífero/metabolismo , Espermatogênese , Espermatozoides/metabolismo , Animais , Humanos , Células Intersticiais do Testículo/metabolismo , Masculino , Células de Sertoli/metabolismo , Transdução de Sinais
13.
Gene ; 780: 145531, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33631249

RESUMO

Sertoli cells are "nurturing cells'' in the seminiferous tubules of the testis which have essential roles in the development, proliferation and differentiation of germ cells. These cells also divide the seminiferous epithelium into a basal and an adluminal compartment and establish the blood-testis barrier (BTB). BTB shields haploid germ cells from recognition by the innate immune system. Moreover, after translocation of germ cells into the adluminal compartment their nutritional source is separated from the circulatory system being only supplied by the Sertoli cells. The integrity of BTB is influenced by several organic/ organometallic, hormonal and inflammatory substances. Moreover, several environmental contaminants such as BPA have hazardous effects on the integrity of BTB. In the current review, we summarize the results of studies that assessed the impact of these agents on the integrity of BTB. These studies have implications in understanding the molecular mechanism of male infertility and also in the male contraception.


Assuntos
Barreira Hematotesticular/metabolismo , Exposição Ambiental/efeitos adversos , Poluentes Ambientais/toxicidade , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/efeitos dos fármacos , Animais , Barreira Hematotesticular/patologia , Humanos , Masculino , Epitélio Seminífero/patologia , Células de Sertoli/patologia
14.
Tissue Cell ; 69: 101484, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33450652

RESUMO

The Sand rat, Psammomys obesus, living northwest of the Algerian Sahara, presents a seasonal reproductive cycle. The purposes of this study were firstly to determine the stages of seminiferous epithelium cycle (SEC) by histological and morphometric analysis and secondly to investigate, for the first time, the testicular expression of RFamide-related peptide-3 (RFRP-3) during the SEC by immunohistochemistry. The results showed that the SEC consists of 14 stages according to the tubular morphology method. RFRP-3 was observed in both testicular compartments: the tubular and the interstitial. Leydig cells exhibited the highest RFRP-3 signal (30.73 % ± 4.80) compared to Sertoli cells (13-15 %). In the germline, RFRP-3 was detected during the late prophase I of meiosis in late pachytene, diplotene and metaphasic spermatocytes I. In addition, only round and triangular spermatids were positive during spermiogenesis. Referring to the SEC, it was found that the increased staining of RFRP-3 in spermatocytes I coincided with late pachytene of XI and XII stages (16.90 % ± 0.69 and 16.61 % ± 0.28, respectively). In spermatids, the labeling decreased in the triangular ones at stage IX (8.04 % ± 0.42). These results suggest the involvement of RFRP-3 in the control of SEC in P. obesus.


Assuntos
Gerbillinae/metabolismo , Neuropeptídeos/metabolismo , Epitélio Seminífero/metabolismo , Animais , Imuno-Histoquímica , Masculino , Epitélio Seminífero/citologia , Testículo/citologia , Testículo/metabolismo
15.
Asian J Androl ; 23(2): 123-128, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32896837

RESUMO

Collagen α3 (IV) chains are one of the major constituent components of the basement membrane in the mammalian testis. Studies have shown that biologically active fragments, such as noncollagenase domain (NC1)-peptide, can be released from the C-terminal region of collagen α3 (IV) chains, possibly through the proteolytic action of metalloproteinase 9 (MMP9). NC1-peptide was shown to promote blood-testis barrier (BTB) remodeling and fully developed spermatid (e.g., sperm) release from the seminiferous epithelium because this bioactive peptide was capable of perturbing the organization of both actin- and microtubule (MT)-based cytoskeletons at the Sertoli cell-cell and also Sertoli-spermatid interface, the ultrastructure known as the basal ectoplasmic specialization (ES) and apical ES, respectively. More importantly, recent studies have shown that this NC1-peptide-induced effects on cytoskeletal organization in the testis are mediated through an activation of mammalian target of rapamycin complex 1/ribosomal protein S6/transforming retrovirus Akt1/2 protein (mTORC1/rpS6/Akt1/2) signaling cascade, involving an activation of cell division control protein 42 homolog (Cdc42) GTPase, but not Ras homolog family member A GTPase (RhoA), and the participation of end-binding protein 1 (EB1), a microtubule plus (+) end tracking protein (+TIP), downstream. Herein, we critically evaluate these findings, providing a critical discussion by which the basement membrane modulates spermatogenesis through one of its locally generated regulatory peptides in the testis.


Assuntos
Membrana Basal/metabolismo , Barreira Hematotesticular/metabolismo , Colágeno Tipo IV/metabolismo , Fragmentos de Peptídeos/metabolismo , Epitélio Seminífero/metabolismo , Espermatogênese/fisiologia , Citoesqueleto de Actina , Animais , Membrana Basal/fisiologia , Barreira Hematotesticular/fisiologia , Comunicação Celular , Colágeno Tipo IV/fisiologia , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos , Fragmentos de Peptídeos/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína S6 Ribossômica/metabolismo , Epitélio Seminífero/fisiologia , Células de Sertoli/metabolismo , Células de Sertoli/fisiologia , Transdução de Sinais , Espermátides/metabolismo , Espermátides/fisiologia , Testículo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
16.
Biol Reprod ; 103(2): 378-389, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32678439

RESUMO

Spermiation is a multiple-step process involving profound cellular changes in both spermatids and Sertoli cells. We have observed spermiation defects, including abnormalities in spermatid orientation, translocation and release, in mice deficient in the retinoic acid receptor alpha (RARA) and upon treatment with a pan-RAR antagonist. To elucidate the role of retinoid signaling in regulating spermiation, we first characterized the time course of appearance of spermiogenic defects in response to treatment with the pan-RAR antagonist. The results revealed that defects in spermiation are indeed among the earliest abnormalities in spermatogenesis observed upon inhibition of retinoid signaling. Using fluorescent dye-conjugated phalloidin to label the ectoplasmic specialization (ES), we showed for the first time that these defects involved improper formation of filamentous actin (F-actin) bundles in step 8-9 spermatids and a failure of the actin-surrounded spermatids to move apically to the lumen and to disassemble the ES. The aberrant F-actin organization is associated with diminished nectin-3 expression in both RARA-deficient and pan-RAR antagonist-treated testes. An abnormal localization of both tyrosinated and detyrosinated tubulins was also observed during spermatid translocation in the seminiferous epithelium in drug-treated testes. These results highlight a crucial role of RAR receptor-mediated retinoid signaling in regulating microtubules and actin dynamics in the cytoskeleton rearrangements, required for proper spermiation. This is critical to understand in light of ongoing efforts to inhibit retinoid signaling as a novel approach for male contraception and may reveal spermiation components that could also be considered as new targets for male contraception.


Assuntos
Actinas/metabolismo , Retinoides/metabolismo , Transdução de Sinais/fisiologia , Espermátides/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Masculino , Camundongos , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo
17.
Reproduction ; 160(2): 205-215, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32438343

RESUMO

The existence of cytoplasmic passages between germ cells and their potential function in the control of the spermatogenic process has long been an intriguing question. Evidence of the important role of such structures, known as intercellular bridges (ICB), in spermatogenesis has been implicated by the failure of spermatogenesis in testis-expressed gene 14 (Tex14) mutant mice, which lack the ICBs, to progress past the pachytene spermatocyte stage. Using these Tex14 mutants, the present study evaluated, for the first time, the behavior and synchrony of the spermatogonial lineage in the absence of ICBs. Our data suggest that the absence of these cytoplasmic connections between cells affects the expansion of the undifferentiated type A (Aundiff) spermatogonia compartment and their transition to A1, resulting in a significant numerical reduction of differentiating A1 spermatogonia, but did not interfere with cell amplification during subsequent mitotic steps of differentiating spermatogonia from A1 through intermediate (In). However, beginning at the type B spermatogonia, the synchrony of differentiation was impaired as some cells showed delayed differentiation compared to their behavior in a normal seminiferous epithelium cycle. Thus although spermatogonial development is able to proceed, in the absence of ICBs in Tex14-/- mutants, the yield of cells, specific steps of differentiation, the synchrony of the cell kinetics, and the subsequent progression in meiosis are quantitatively lower than normal.


Assuntos
Comunicação Celular , Diferenciação Celular , Meiose , Epitélio Seminífero/patologia , Espermatogênese , Espermatogônias/patologia , Fatores de Transcrição/fisiologia , Animais , Proliferação de Células , Citoplasma , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Epitélio Seminífero/metabolismo , Espermatogônias/metabolismo
18.
Reprod Biol Endocrinol ; 18(1): 30, 2020 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-32299422

RESUMO

BACKGROUND: Onset of spermatogenesis at puberty is critically dependent on the activity of hypothalamic-pituitary-gonadal axis and testosterone production by Leydig cells. The aim of this study was to examine whether activation of Notch receptors and expression of Notch ligands and effector genes in rat seminiferous epithelium are controlled by androgen signaling during puberty. METHODS: Peripubertal (5-week-old) Wistar rats received injections of flutamide (50 mg/kg bw) daily for 7 days to reduce androgen receptor (AR) signaling or a single injection of ethanedimethane sulphonate (EDS; 75 mg/kg bw) to reduce testosterone production. Gene and protein expressions were analyzed by real-time RT-PCR and western blotting, respectively, protein distribution by immunohistochemistry, and steroid hormone concentrations by enzyme-linked immunosorbent assay. Statistical analyses were performed using one-way ANOVA followed by Tukey's post hoc test or by Kruskal-Wallis test, followed by Dunn's test. RESULTS: In both experimental models changes of a similar nature in the expression of Notch pathway components were found. Androgen deprivation caused the reduction of mRNA and protein expression of DLL4 ligand, activated forms of Notch1 and Notch2 receptors and HES1 and HEY1 effector genes (p < 0.05, p < 0.01, p < 0.001). In contrast, DLL1, JAG1 and HES5 expressions increased in seminiferous epithelium of both flutamide and EDS-treated rats (p < 0.05, p < 0.01, p < 0.001). CONCLUSIONS: Androgens and androgen receptor signaling may be considered as factors regulating Notch pathway activity and the expression of Hes and Hey genes in rat seminiferous epithelium during pubertal development. Further studies should focus on functional significance of androgen-Notch signaling cross-talk in the initiation and maintenance of spermatogenesis.


Assuntos
Flutamida/farmacologia , Receptores Androgênicos/metabolismo , Receptores Notch/metabolismo , Epitélio Seminífero/efeitos dos fármacos , Maturidade Sexual/fisiologia , Transdução de Sinais/efeitos dos fármacos , Antagonistas de Androgênios/administração & dosagem , Antagonistas de Androgênios/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Flutamida/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Masculino , Ratos Wistar , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Epitélio Seminífero/metabolismo , Testosterona/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
19.
Mol Reprod Dev ; 87(4): 419-429, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32020743

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) and retinoic acid (RA) are two molecules crucial for the regulation of the spermatogonial compartment of the testis. During the cycle of the seminiferous epithelium, their relative concentration oscillates with lower GDNF levels in stages where RA levels are high. It has been recently shown that RA negatively regulates Gdnf expression but the mechanisms behind are so far unknown. Here, we show that RA directly downregulates Gdnf mRNA levels in primary murine Sertoli cells through binding of RARα to a novel DR5-RARE on Gdnf promoter. Pharmacological inhibition and chromatin immunoprecipitation-quantitative polymerase chain reaction analysis suggested that the underlying mechanism involved histone deacetylase activity and epigenetic repression of Gdnf promoter upon RA treatment.


Assuntos
Regulação para Baixo/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células de Sertoli/metabolismo , Tretinoína/metabolismo , Tretinoína/farmacologia , Animais , Benzoatos/farmacologia , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Masculino , Camundongos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Espermatogônias/metabolismo , Estilbenos/farmacologia , Transfecção
20.
Reprod Sci ; 27(7): 1443-1454, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31994000

RESUMO

Sertoli cells are important for spermatogenesis not only by directly interacting with germ line cells in the seminiferous epithelium but also by constituting the blood-testis barrier (BTB) structure to create a favorable environment for spermatogenesis. Blind sterile (bs) male mice are infertile, with excessive germ cell apoptosis and spermatogenesis arrest. TBC1D20 (TBC1 domain family member 20) deficiency has been identified as the causative mutation in bs mice. However, whether TBC1D20 loss of function also impairs BTB integrity, which further contributes to the failed spermatogenesis of bs male mice, remains unclear. In the present study, biotin tracer assay and transmission electron microscopy showed severely disrupted BTB integrity in bs testes. Compared to the wild-type Sertoli cells, BTB components of cultured bs Sertoli cells in vitro was perturbed with downregulation of E-cadherin, ZO-1, ß-catenin, and Claudin 11. The obvious rearrangement of F-actin indicated disrupted epithelial-mesenchymal balance in TBC1D20-deficient Sertoli cells. The ability of bs Sertoli cells to maintain the clone formation of spermatogonia stem cells was also obviously limited. Furthermore, the decreasing of SOX9 (sex-determining region Y box 9) and WT1 (Wilms' tumor 1) and increasing of vimentin in bs Sertoli cells indicated that TBC1D20 loss of function attenuated the differentiation progression of bs Sertoli cells. In summary, TBC1D20 loss of function impedes the maturation of adult Sertoli cells and resulted in impaired BTB integrity, which is further implicated in the infertile phenotype of bs male mice.


Assuntos
Barreira Hematotesticular/metabolismo , Epitélio Seminífero/metabolismo , Células de Sertoli/metabolismo , Proteínas rab1 de Ligação ao GTP/efeitos dos fármacos , Animais , Barreira Hematotesticular/patologia , Células Cultivadas , Técnicas de Cocultura , Infertilidade Masculina/genética , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Transgênicos , Epitélio Seminífero/patologia , Células de Sertoli/patologia , Testículo/metabolismo , Testículo/patologia , Proteínas rab1 de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...