Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.243
Filtrar
1.
Reprod Toxicol ; 123: 108497, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37949197

RESUMEN

Diuron is a phenylurea herbicide widely used in the agricultural industry. In recent years, the risk of infertility and developmental defects has increased due to exposure to environmental pollutants. In this study, we investigated the toxicity of diuron in fetal mouse testes using three-dimensional organ cultures. Fetal testes derived from embryonic day (E) 14.5 were cultured with 200 µM diuron for 5 days. The results revealed that diuron did not impair fetal germ cell proliferation or the expression levels of germ cell markers such as Ddx4, Dazl, Oct 4, Nanog, Plzf, and TRA 98. Similarly, the gene or protein expression of the Sertoli cell markers Sox9 and Wt1 in diuron-exposed fetal testes did not change after 5 days of culture. In contrast, diuron increased fetal Leydig cell markers (FLC), Cyp11a1, Cyp17a1, Thbs2, and Pdgf α, and decreased adult Leydig cell (ALC) markers, Sult1e1, Hsd173, Ptgds, and Vcam1. However, 3-ßHSD, an FLC and ALC marker, was consistently maintained upon exposure to diuron in fetal testes compared to non-treated groups. In conclusion, our study demonstrates that diuron negatively impacts Fetal Leydig cell development, although it does not affect germ and Sertoli cells.


Asunto(s)
Células Intersticiales del Testículo , Testículo , Ratones , Masculino , Animales , Testículo/metabolismo , Células Intersticiales del Testículo/metabolismo , Diurona/metabolismo , Células de Sertoli/fisiología , Feto/metabolismo
2.
Front Endocrinol (Lausanne) ; 14: 1110572, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37124741

RESUMEN

Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.


Asunto(s)
Espermatogénesis , Testículo , Masculino , Ratones , Animales , Espermatogénesis/fisiología , Células de Sertoli/fisiología , Gonadotropinas , Hormona Folículo Estimulante/fisiología , Hormona Luteinizante , Mamíferos
3.
Int J Mol Sci ; 24(2)2023 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-36674801

RESUMEN

Canonical coxsackievirus and adenovirus receptor (CXADR) is a transmembrane component of cell junctions that is crucial for cardiac and testicular functions via its homophilic and heterophilic interaction. CXADR is expressed in both Sertoli cells and germ cells and is localized mainly at the interface between Sertoli-Sertoli cells and Sertoli-germ cells. Knockout of CXADR in mouse Sertoli cells specifically impairs male reproductive functions, including a compromised blood-testis barrier, apoptosis of germ cells, and premature loss of spermatids. Apart from serving as an important component for cell junctions, recent progress has showed the potential roles of CXADR as a signaling mediator in spermatogenesis. This review summarizes current research progress related to the regulation and role of CXADR in spermatogenesis as well as in pathological conditions. We hope this review provides some future directions and a blueprint to promote the further study on the roles of CXADR.


Asunto(s)
Receptores Virales , Espermatogénesis , Animales , Masculino , Ratones , Infecciones por Coxsackievirus , Enterovirus , Ratones Noqueados , Receptores Virales/metabolismo , Células de Sertoli/fisiología , Espermátides , Testículo
4.
Andrology ; 11(5): 849-859, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36651593

RESUMEN

Sertoli cells are somatic cells that are in close contact with germ cells in the mammalian testes. They have multiple functions and fulfill key roles for the development and proper maturation of spermatogenic cells into functional spermatozoa. One of their most important properties is to release trophic factors and supply nutrients to germ cells. But they are also involved in the regulation of the immune system in testis, and provide an immunologically privileged environment for developing germ cells. Because they are so essential for reproductive cells, their alterations can have detrimental consequences for fertility. Many environmental factors and exposures such as high caloric diet, toxins, and pollutants are thought to compromise Sertoli cells physiology. This review describes the discovery of Sertoli cells and the methods used for their study, summarizes their major properties and functions, and describes their dysfunctions in pathologies, particularly associated with environmental stressors.


Asunto(s)
Células de Sertoli , Testículo , Masculino , Animales , Células de Sertoli/fisiología , Testículo/fisiología , Espermatozoides , Células Germinativas , Fertilidad , Espermatogénesis/fisiología , Mamíferos
5.
Cell Tissue Res ; 392(2): 581-604, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36627392

RESUMEN

Experimental autoimmune orchitis (EAO) is a chronic inflammatory disorder that causes progressive spermatogenic impairment. EAO is characterized by high intratesticular levels of nitric oxide (NO) and tumor necrosis factor alpha (TNFα) causing germ cell apoptosis and Sertoli cell dysfunction. However, the impact of this inflammatory milieu on the spermatogenic wave is unknown. Therefore, we studied the effect of inflammation on spermatogonia and preleptotene spermatocyte cell cycle progression in an EAO context and through the intratesticular DETA-NO and TNFα injection in the normal rat testes. In EAO, premeiotic germ cell proliferation is limited as a consequence of the undifferentiated spermatogonia (CD9+) cell cycle arrest in G2/M and the reduced number of differentiated spermatogonia (c-kit+) and preleptotene spermatocytes that enter in the meiotic S-phase. Although inflammation disrupts spermatogenesis in EAO, it is maintained in some seminiferous tubules at XIV and VII-VIII stages of the epithelial cell cycle, thereby guaranteeing sperm production. We found that DETA-NO (2 mM) injected in normal testes arrests spermatogonia and preleptotene spermatocyte cell cycle; this effect reduces the number of proliferative spermatogonia and the number of preleptotene spermatocytes in meiosis S-phase (36 h after). The temporal inhibition of spermatogonia clonal amplification delayed progression of the spermatogenic wave (5 days after) finally altering spermatogenesis. TNFα (0.5 and 1 µg) exposure did not affect premeiotic germ cell cycle or spermatogenic wave. Our results show that in EAO the inflammatory microenvironment altered spermatogenesis kinetics through premeiotic germ cell cycle arrest and that NO is a sufficient factor contributing to this phenomenon.


Asunto(s)
Orquitis , Factor de Necrosis Tumoral alfa , Ratas , Humanos , Animales , Masculino , Factor de Necrosis Tumoral alfa/farmacología , Semen , Espermatogénesis/fisiología , Espermatogonias , Testículo , Espermatocitos , Células de Sertoli/fisiología , Inflamación/patología
6.
Theriogenology ; 189: 20-30, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35714522

RESUMEN

Elevated glucocorticoid (GC) concentrations associated with captivity-related stress have been linked to impaired testicular function and low sperm quality in felids, but direct physiological evidence is lacking. This study assessed the effects of exogenous GC treatment on felid testicular function using the domestic cat (Felis catus) as a model species. Sixteen intact male cats aged 2.4 ± 0.8 years (mean ± SEM) were divided randomly into treatment (n = 8) and control (n = 8) groups. Treatment cats were given 1 mg kg-1 oral prednisolone daily for 50 days. Blood samples were taken on Days 0 (first prednisolone treatment), 2, 4, 7, 10, 20, 30, 40, 50 (prior to neutering) and 60 of the trial. All cats were orchiectomised on day 50, epididymal sperm assessed, and the testes fixed for histological assessment. Testosterone concentrations did not differ between the two groups. While sperm motility was similar between the treatment and control groups, cats given prednisolone had a higher proportion of morphologically abnormal sperm in both the caput (72.5% vs. 59.6%, P < 0.001) and cauda (56.7% vs. 35.8%, P < 0.001) epididymis. Testicular histomorphometric data and total number of germ cells per seminiferous tubule cross section did not differ between groups, nor did the relative abundance of spermatogonia, spermatocytes, and spermatids. Cats given prednisolone had fewer Sertoli cells per tubule cross-section than those in the control group (17.1 ± 0.9 vs. 19.7 ± 0.8, P = 0.04), which was likely related to higher rates of Sertoli cell apoptosis in treatment versus control cats (0.25 ± 0.02 vs. 0.10 ± 0.02 apoptotic Sertoli cells per tubule, respectively; P < 0.001). Sertoli cell load (number of germ cells per Sertoli cell) was also higher in the treatment group than in the control group (11.5 ± 0.8 vs. 9.4 ± 1.2 germ cells per Sertoli cell, respectively; P < 0.001), and was positively correlated with the percentage of morphologically abnormal sperm in the epididymis (r2 = 0.78, P < 0.001). Prednisolone treatment resulted in an increase in the proportion of abnormal sperm in the epididymis, which may be explained by an increased nurturing demand on a reduced Sertoli cell population. These findings provide novel evidence to support the hypothesis that elevated GC concentrations, such as those resulting from captivity-related stress, have the potential to impair testicular function and sperm quality in felids.


Asunto(s)
Epidídimo , Células de Sertoli , Animales , Gatos , Epidídimo/fisiología , Glucocorticoides/farmacología , Masculino , Prednisolona/farmacología , Semen , Células de Sertoli/fisiología , Motilidad Espermática/fisiología , Espermátides , Espermatogénesis/fisiología , Espermatozoides/fisiología , Testículo
7.
Reproduction ; 164(1): 9-18, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35521906

RESUMEN

Spermatozoa released from Sertoli cells must be transported to the epididymis. However, the mechanism of the luminal flow in seminiferous tubules has remained unclear to date. Therefore, in this study, we investigated luminal flow and movements in the seminiferous tubules by three-dimensional analysis and in vivo imaging. Serial 5-µm-thick mouse testicular sections at 50-µm-intervals were prepared and stained by Periodic Acid-Schiff-hematoxylin. After three-dimensional reconstruction of the seminiferous tubules, the localization of the released spermatozoa and the stages observed in the sections were recorded in each reconstructed tubule. Luminal movements in the seminiferous tubules were observed by in vivo imaging using a fluorescent-reporter mouse and two-photon excitation microscopy system. Spermatozoa without contact to the seminiferous epithelium were not accumulated toward the rete testis. Additionally, such spermatozoa were found on their way not only to the most proximal rete testis but also a more distant rete testis from any stage VIII seminiferous epithelia. In vivo imaging demonstrated that the direction of the flagella of spermatozoa attached to the seminiferous epithelium was repeatedly reversed. The epithelium at the inner curve of the seminiferous tubule was shaken more actively and had fewer spermatozoa attached compared with the epithelium at the outer curve. Our results hence suggest that the luminal flow in the seminiferous tubules is repeatedly reversed and that this physical force helps spermatozoa to be released from Sertoli cells. In brief: Spermatozoa are released from Sertoli cells and flow in the seminiferous tubule to the rete testis. Our results suggest that the luminal flow in the tubules is repeatedly reversed and that this physical force helps spermatozoa release from the Sertoli cells.


Asunto(s)
Microfluídica , Túbulos Seminíferos , Células de Sertoli , Espermatozoides , Animales , Imagenología Tridimensional , Masculino , Ratones , Microfluídica/métodos , Microscopía , Red Testicular/fisiología , Reología/métodos , Epitelio Seminífero/diagnóstico por imagen , Epitelio Seminífero/fisiología , Túbulos Seminíferos/diagnóstico por imagen , Túbulos Seminíferos/fisiología , Células de Sertoli/fisiología , Espermatozoides/fisiología , Testículo/diagnóstico por imagen , Testículo/fisiología
8.
Theriogenology ; 188: 13-21, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35636243

RESUMEN

Somatic Sertoli cells constitute the microenvironment and produce essential substances, to support male germ cell development and maturation in testis. We previously found that ascorbic acid treatment of porcine immature Sertoli cells enhances its proliferation and secretion of reproductive hormones and metabolites, and reprograms the global transcriptome. Proteomics is a powerful tool to systematically profile the underlying protein changes. Here, by employing the TMT-based quantitative proteomics method, we identified 96 and 64 significantly up- and down-regulated proteins in porcine immature Sertoli cells treated by ascorbic acid, respectively. Gene enrichment (GO and KEGG) and protein-protein interaction analyses revealed important molecular pathways (dioxygenase activity, sterol biosynthetic process, PI3K-Akt, negative regulation of peptide hormone secretion, extracellular matrix etc.). Further validation of three proteins, HMGCS1 (cholesterol synthesis), P4HA1 (glycolysis) and KDM5A (demethylation of histone 3 at lysine 4), confirmed their significant differential abundance, respectively. Taken together, our findings show that ascorbic acid can alter multiple important protein molecules and related signaling pathways, which could explain partially phenotypic changes (proliferation, apoptosis, nucleic acid methylation, lactate and reproductive hormone secretion) of porcine immature Sertoli cells as induced by ascorbic acid.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Células de Sertoli , Animales , Ácido Ascórbico/farmacología , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Proteómica , Células de Sertoli/fisiología , Porcinos , Testículo/metabolismo
9.
Mol Hum Reprod ; 28(2)2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35088858

RESUMEN

Reciprocal communication between Sertoli and Leydig cells occurs in the testes; however, the detailed mechanisms involved are not completely understood. Exosomes can communicate within neighboring or distant cells to regulate cell function. Our aim was to determine whether exosomes released from Sertoli cells can regulate the survival of Leydig cells. We found that exosomes released from rat primary Sertoli cells could be internalized by Leydig cells in vitro, and promote the survival of Leydig cells, as assessed by optical density at 450 nm, compared to untreated control (mean ± SD: 0.95 ± 0.04 vs 0.79 ± 0.03, P < 0.05). When the exosomes were injected into the interstitial area of rat testis, they could also be internalized by Leydig cells in vivo. To investigate if exosomes released from Sertoli cells can reach Leydig cells in vivo, exosomes were injected into the efferent duct, from where they entered the interstitial space from seminiferous tubules, which indicated that they may cross the blood-testis barrier (BTB). Further in vitro studies found that exosomes released from Sertoli cells significantly increased CC-chemokine ligand 20 (Ccl20) mRNA (mean ± SD: 2.79 ± 0.08 vs 0.98 ± 0.04, P < 0.01) and protein (mean ± SD: 1.08 ± 0.06 vs 0.53 ± 0.05 ng/ml, P < 0.01) levels in Leydig cells, compared to the untreated Leydig cells. CCL20 promoted the phosphorylation of AKT (protein kinase B) in Leydig cells, compared to untreated control (mean ± SD: 0.074 ± 0.002 vs 0.051 ± 0.002, P < 0.01). In conclusion, our results demonstrated that exosomes released by Sertoli cells may cross the BTB and promote the survival of Leydig cells. The findings may add new evidence for Sertoli-Leydig cell communication.


Asunto(s)
Exosomas , Células de Sertoli , Animales , Células Intersticiales del Testículo , Masculino , Ratas , Túbulos Seminíferos , Células de Sertoli/fisiología , Testículo/fisiología
10.
FASEB J ; 36(2): e22131, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34985827

RESUMEN

Although germ cell fate is believed to be determined by signaling factors from differentiated somatic cells, the molecular mechanism behind this process remains obscure. In this study, premature meiosis in male germ cells was observed during the embryonic stage by conditional activation of ß-catenin in Sertoli cells. Somatic and germ cell transcriptome results indicated that the BMP signaling pathway was enriched after ß-catenin activation. In addition, we observed a decreased DNA methylation within a reduction of DNMT3A in germ cells of ß-catenin activated testes and reversed increase after inhibiting BMP signaling pathway with LDN-193189. We also found that Dazl expression was increased in ß-catenin activated testes and decreased after LDN treatment. Taken together, this study demonstrates that male germ cells entered meiosis prematurely during the embryonic stage after ß-catenin activated in Sertoli cells. BMP signaling pathway involved in germ cell meiosis initiation by mediating DNA methylation to induce meiotic genes expression.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Desarrollo Embrionario/genética , Células Germinativas/fisiología , Meiosis/genética , Proteínas de Unión al ARN/genética , Regulación hacia Arriba/genética , Animales , Diferenciación Celular/genética , Metilación de ADN/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Células de Sertoli/fisiología , Transducción de Señal/genética , Testículo/patología , Transcriptoma/genética , beta Catenina/genética
11.
Front Endocrinol (Lausanne) ; 12: 761249, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721307

RESUMEN

To achieve spermatogenesis in vitro, one of the most challenging processes to mimic is meiosis. Meiotic problems, like incomplete synapsis of the homologous chromosomes, or impaired homologous recombination, can cause failure of crossover formation and subsequent chromosome nondisjunction, eventually leading to aneuploid sperm. These meiotic events are therefore strictly monitored by meiotic checkpoints that initiate apoptosis of aberrant spermatocytes and lead to spermatogenic arrest. However, we recently found that, in vitro derived meiotic cells proceeded to the first meiotic division (MI) stage, despite displaying incomplete chromosome synapsis, no discernible XY-body and lack of crossover formation. We therefore optimized our in vitro culture system of meiosis from male germline stem cells (mGSCs) in order to achieve full chromosome synapsis, XY-body formation and meiotic crossovers. In comparison to previous culture system, the in vitro-generated spermatocytes were transferred after meiotic initiation to a second culture dish. This dish already contained a freshly plated monolayer of proliferatively inactivated immortalized Sertoli cells supporting undifferentiated mGSCs. In this way we aimed to simulate the multiple layers of germ cell types that support spermatogenesis in vivo in the testis. We found that in this optimized culture system, although independent of the undifferentiated mGSCs, meiotic chromosome synapsis was complete and XY body appeared normal. However, meiotic recombination still occurred insufficiently and only few meiotic crossovers were formed, leading to MI-spermatocytes displaying univalent chromosomes (paired sister chromatids). Therefore, considering that meiotic checkpoints are not necessarily fully functional in vitro, meiotic crossover formation should be closely monitored when mimicking gametogenesis in vitro to prevent generation of aneuploid gametes.


Asunto(s)
Emparejamiento Cromosómico/fisiología , Cromosomas/fisiología , Meiosis/fisiología , Aneuploidia , Animales , Azoospermia/congénito , Azoospermia/fisiopatología , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Células de Sertoli/fisiología , Espermatocitos/fisiología , Espermatogénesis/fisiología , Espermatozoides/fisiología , Testículo/fisiología
12.
Genes (Basel) ; 12(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34573441

RESUMEN

As in other vertebrates, avian testes are the site of spermatogenesis and androgen production. The paired testes of birds differentiate during embryogenesis, first marked by the development of pre-Sertoli cells in the gonadal primordium and their condensation into seminiferous cords. Germ cells become enclosed in these cords and enter mitotic arrest, while steroidogenic Leydig cells subsequently differentiate around the cords. This review describes our current understanding of avian testis development at the cell biology and genetic levels. Most of this knowledge has come from studies on the chicken embryo, though other species are increasingly being examined. In chicken, testis development is governed by the Z-chromosome-linked DMRT1 gene, which directly or indirectly activates the male factors, HEMGN, SOX9 and AMH. Recent single cell RNA-seq has defined cell lineage specification during chicken testis development, while comparative studies point to deep conservation of avian testis formation. Lastly, we identify areas of future research on the genetics of avian testis development.


Asunto(s)
Aves , Células de Sertoli/citología , Testículo/anatomía & histología , Testículo/fisiología , Animales , Aves/anatomía & histología , Aves/fisiología , Diferenciación Celular/genética , Pollos , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Masculino , Células de Sertoli/fisiología , Procesos de Determinación del Sexo , Testículo/embriología , Testículo/crecimiento & desarrollo , Factores de Transcripción/genética
13.
Biol Reprod ; 105(5): 1344-1354, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34467369

RESUMEN

Seasonal reproduction contributes to increased chances of offspring survival in some animals. Dairy goats are seasonal breeding mammals. In this study, adult male Guanzhong dairy goats (10-12 months old) were used. Testis size, semen quality, hormone level, apoptosis of germ cells, and autophagy of Sertoli cells were analyzed in dairy goats during the breeding (October) and nonbreeding (April) seasons. We found that, during the nonbreeding season for dairy goats, semen quality, follicle-stimulating hormone (FSH) levels, and testosterone levels were reduced, and the number of apoptotic germ cells increased. The proliferation with decrease activity of germ cells in dairy goat during the nonbreeding season was significantly affected. However, the testis size did not change seasonally. Interestingly, Sertoli cell autophagy was more active during the nonbreeding season. The expression levels of FSH receptor, wilms tumor 1, androgen binding protein, glial cell derived neurotrophic factor, and stem cell factor decreased in dairy goats during the nonbreeding season. In summary, our results indicate that spermatogenesis in dairy goats during the nonbreeding season was not completely arrested. In addition, germ cell apoptosis and the morphology of Sertoli cells considerably changed in dairy goats during the nonbreeding season. Sertoli cell autophagy is involved in the seasonal regulation of spermatogenesis in dairy goats. These findings provide key insights into the fertility and spermatogenesis of seasonal breeding animals.


Asunto(s)
Autofagia , Expresión Génica , Testículo/fisiología , Animales , Cabras/anatomía & histología , Cabras/genética , Cabras/fisiología , Masculino , Estaciones del Año , Análisis de Semen , Células de Sertoli/fisiología , Testículo/anatomía & histología
14.
PLoS Genet ; 17(8): e1009688, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34351902

RESUMEN

Autophagy degrades unnecessary proteins or damaged organelles to maintain cellular function. Therefore, autophagy has a preventive role against various diseases including hepatic disorders, neurodegenerative diseases, and cancer. Although autophagy in germ cells or Sertoli cells is known to be required for spermatogenesis and male fertility, it remains poorly understood how autophagy participates in spermatogenesis. We found that systemic knockout mice of Rubicon, a negative regulator of autophagy, exhibited a substantial reduction in testicular weight, spermatogenesis, and male fertility, associated with upregulation of autophagy. Rubicon-null mice also had lower levels of mRNAs of Sertoli cell-related genes in testis. Importantly, Rubicon knockout in Sertoli cells, but not in germ cells, caused a defect in spermatogenesis and germline stem cell maintenance in mice, indicating a critical role of Rubicon in Sertoli cells. In mechanistic terms, genetic loss of Rubicon promoted autophagic degradation of GATA4, a transcription factor that is essential for Sertoli cell function. Furthermore, androgen antagonists caused a significant decrease in the levels of Rubicon and GATA4 in testis, accompanied by elevated autophagy. Collectively, we propose that Rubicon promotes Sertoli cell function by preventing autophagic degradation of GATA4, and that this mechanism could be regulated by androgens.


Asunto(s)
Factor de Transcripción GATA4/metabolismo , Técnicas de Inactivación de Genes/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Células de Sertoli/fisiología , Animales , Autofagia , Línea Celular , Fertilidad , Humanos , Masculino , Ratones , Proteolisis , Células de Sertoli/citología , Análisis de la Célula Individual , Espermatogénesis , Testículo/crecimiento & desarrollo , Testículo/metabolismo
15.
Mol Hum Reprod ; 27(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34273154

RESUMEN

Sertoli cells provide protection and nutrition for developing sperm. Each stage of sperm development occurs on the surface of Sertoli cells. MicroRNA (MiR)-125a-5p is involved in male reproduction. The current research aimed to probe the role of miR-125a-5p in Sertoli cell function. Functionally, miR-125a-5p knockdown facilitated Sertoli cell proliferation, while miR-125a-5p overexpression suppressed Sertoli cell proliferation, as evidenced by 5-ethynyl-20-deoxyuridine incorporation assay. Additionally, miR-125a-5p knockdown inhibited Sertoli cell apoptosis, while miR-125a-5p upregulation facilitated Sertoli cell apoptosis, as evidenced by flow cytometry analysis. Computationally, we identified four predicted mRNA targets of miR-125a-5p. Based on the results of luciferase reporter assay, miR-125a-5p was confirmed to bind to the predicted sequence in the Ras-related protein Rab-3D (RAB3D) 3'UTR. Rescue experiments showed that miR-125a-5p suppressed the proliferative ability of TM4 Sertoli cells and facilitated their apoptosis by targeting RAB3D. Finally, our data confirmed that miR-125a-5p and RAB3D modulated activation of the PI3K/AKT pathway. In conclusion, our data showed that miR-125a-5p regulated Sertoli cell proliferation and apoptosis by targeting RAB3D and regulating the PI3K/AKT pathway.


Asunto(s)
MicroARNs/fisiología , Células de Sertoli/fisiología , Proteínas de Unión al GTP rab3/genética , Animales , Apoptosis/genética , Línea Celular , Proliferación Celular/genética , Regulación de la Expresión Génica , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética
16.
Front Endocrinol (Lausanne) ; 12: 694796, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093450

RESUMEN

The incidence of cancer in pre-pubertal boys has significantly increased and, it has been recognized that the gonado-toxic effect of the cancer treatments may lead to infertility. Here, we have evaluated the effects on porcine neonatal Sertoli cells (SCs) of three commonly used chemotherapy drugs; cisplatin, 4-Hydroperoxycyclophosphamide and doxorubicin. All three drugs induced a statistical reduction of 5-hydroxymethylcytosine in comparison with the control group, performed by Immunofluorescence Analysis. The gene and protein expression levels of GDNF, were significantly down-regulated after treatment to all three chemotherapy drugs comparison with the control group. Specifically, differences in the mRNA levels of GDNF were: 0,8200 ± 0,0440, 0,6400 ± 0,0140, 0,4400 ± 0,0130 fold change at 0.33, 1.66, and 3.33µM cisplatin concentrations, respectively (**p < 0.01 at 0.33 and 1.66 µM vs SCs and ***p < 0.001 at 3.33µM vs SCs); 0,6000 ± 0,0340, 0,4200 ± 0,0130 fold change at 50 and 100 µM of 4-Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7000 ± 0,0340, 0,6200 ± 0,0240, 0,4000 ± 0,0230 fold change at 0.1, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 µM vs SCs and ***p < 0.001 at 1 µM vs SCs). Differences in the protein expression levels of GDNF were: 0,7400 ± 0,0340, 0,2000 ± 0,0240, 0,0400 ± 0,0230 A.U. at 0.33, 1.66, and 3.33µM cisplatin concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7300 ± 0,0340, 0,4000 ± 0,0130 A.U. at 50 and 100 µM of 4- Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,6200 ± 0,0340, 0,4000 ± 0,0240, 0,3800 ± 0,0230 A.U. at 0.l, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 µM vs SCs and ***p < 0.001 at 1 µM vs SCs). Furthermore, we have demonstrated the protective effect of eicosapentaenoic acid on SCs only at the highest concentration of cisplatin, resulting in an increase in both gene and protein expression levels of GDNF (1,3400 ± 0,0280 fold change; **p < 0.01 vs SCs); and of AMH and inhibin B that were significantly recovered with values comparable to the control group. Results from this study, offers the opportunity to develop future therapeutic strategies for male fertility management, especially in pre-pubertal boys.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ácido Eicosapentaenoico/farmacología , Preservación de la Fertilidad/métodos , Células de Sertoli/efectos de los fármacos , Animales , Animales Recién Nacidos , Supervivientes de Cáncer , Células Cultivadas , Niño , Cisplatino/efectos adversos , Ácido Eicosapentaenoico/uso terapéutico , Fertilidad/efectos de los fármacos , Gónadas/efectos de los fármacos , Gónadas/patología , Humanos , Masculino , Células de Sertoli/citología , Células de Sertoli/fisiología , Porcinos
17.
Cell Death Dis ; 12(6): 604, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117213

RESUMEN

Kindlin-2 is known to play important roles in the development of mesoderm-derived tissues including myocardium, smooth muscle, cartilage and blood vessels. However, nothing is known for the role of Kindlin-2 in mesoderm-derived reproductive organs. Here, we report that loss of Kindlin-2 in Sertoli cells caused severe testis hypoplasia, abnormal germ cell development and complete infertility in male mice. Functionally, loss of Kindlin-2 inhibits proliferation, increases apoptosis, impairs phagocytosis in Sertoli cells and destroyed the integration of blood-testis barrier structure in testes. Mechanistically, Kindlin-2 interacts with LATS1 and YAP, the key components of Hippo pathway. Kindlin-2 impedes LATS1 interaction with YAP, and depletion of Kindlin-2 enhances LATS1 interaction with YAP, increases YAP phosphorylation and decreases its nuclear translocation. For clinical relevance, lower Kindlin-2 expression and decreased nucleus localization of YAP was found in SCOS patients. Collectively, we demonstrated that Kindlin-2 in Sertoli cells is essential for sperm development and male reproduction.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Fertilidad/genética , Proteínas Musculares/fisiología , Testículo/crecimiento & desarrollo , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Células HEK293 , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/fisiología , Testículo/metabolismo
18.
Andrology ; 9(6): 1911-1922, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34128333

RESUMEN

BACKGROUND: Testis cord elongation and coiling, which occur in the final stage of testis formation, have been attributed to Sertoli cell proliferation; however, the underlying mechanisms remain unclear. OBJECTIVE: The aim of the present study was to clarify the precise three-dimensional structure of testis cords in the final stage of testis formation in mice and rats. MATERIALS AND METHODS: We reconstructed whole testis cords in the final stage of testis formation in mice (on embryonic days 15.5 and 18.5) and rats (on embryonic days 16.5 and 19.5) using serial paraffin sections and high-performance three-dimensional reconstruction software. RESULTS: Detailed morphometric parameters were calculated for three-dimensionally reconstructed testis cords in six mouse and rat testes each. The mean numbers of testis cords in mice and rats were 12.7 and 27.8, respectively. The mean number of branching points per testis cord was 1.52 in mice, whereas it was only 0.30 in rats. In contrast, the mean ratio of the inner cords, that is, cords not in contact with the tunica albuginea, was 23.0% in rats, whereas it was only 6.5% in mice. In both species, the cords on the cranial side coiled more strongly than those on the caudal side, consistent with the greater expansion of the testis volume on the caudal side. All cords formed right-handed helices from the rete testis side. DISCUSSION AND CONCLUSIONS: The present results suggest that testis cords undergo anastomosis at a higher frequency in mice than in rats and that the coiling of testis cords proceeds from the cranial to caudal side of the testis in both species.


Asunto(s)
Imagenología Tridimensional , Modelos Anatómicos , Cordón Espermático/embriología , Testículo/embriología , Animales , Proliferación Celular/fisiología , Masculino , Ratones , Modelos Animales , Ratas , Células de Sertoli/fisiología
19.
Mol Reprod Dev ; 88(6): 405-415, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34032349

RESUMEN

The effect of stress on male fertility is a widespread public health issue, but less is known about the related signaling pathway. To investigate this, we established a hypercortisolism mouse model by supplementing the drinking water with corticosterone for four weeks. In the hypercortisolism mice, the serum corticosterone was much higher than in the control, and serum testosterone was significantly decreased. Moreover, corticosterone treatment induced decrease of sperm counts and increase of teratozoospermia. Increased numbers of multinucleated giant cells and apoptotic germ cells as well as downregulated meiotic markers suggested that corticosterone induced impaired spermatogenesis. Further, upregulation of macrophage-specific marker antigen F4/80 as well as inflammation-related genes suggested that corticosterone induced inflammation in the testis. Lactate content was found to be decreased in the testis and Sertoli cells after corticosterone treatment, and lactate metabolism-related genes were downregulated. In vitro phagocytosis assays showed that the phagocytic activity in corticosterone-treated Sertoli cells was downregulated and accompanied by decreased mitochondrial membrane potential, while pyruvate dehydrogenase kinase-4 inhibitor supplementation restored this process. Taken together, our results demonstrated that dysfunctional phagocytosis capacity and lactate metabolism in Sertoli cells participates in corticosterone-induced impairment of spermatogenesis.


Asunto(s)
Glucocorticoides/toxicidad , Células de Sertoli/fisiología , Espermatogénesis/efectos de los fármacos , Animales , Proteínas de Unión al Calcio/análisis , Corticosterona/toxicidad , Síndrome de Cushing/sangre , Síndrome de Cushing/inducido químicamente , Síndrome de Cushing/fisiopatología , Ácido Dicloroacético/farmacología , Hormona Folículo Estimulante/sangre , Ácido Láctico/metabolismo , Hormona Luteinizante/sangre , Masculino , Meiosis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Orquitis/inducido químicamente , Orquitis/metabolismo , Fagocitosis/efectos de los fármacos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/análisis , Células de Sertoli/metabolismo , Recuento de Espermatozoides , Espermatozoides/patología , Testículo/metabolismo , Testosterona/sangre
20.
Expert Rev Mol Diagn ; 21(6): 613-626, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33945376

RESUMEN

INTRODUCTION: Sertoli cells play central roles in the development of testis formation in fetuses and the initiation and maintenance of spermatogenesis in puberty and adulthood, and disorders of Sertoli cell proliferation and/or functional maturation can cause male reproductive disorders at various life stages. It's well documented that various genes are either overexpressed or absent in Sertoli cells during the conversion of an immature, proliferating Sertoli cell to a mature, non-proliferating Sertoli cell, which are considered as Sertoli cell stage-specific markers. Thus, it is paramount to choose an appropriate Sertoli cell marker that will be used not only to identify the developmental, proliferative, and maturation of Sertoli cell status in the testis during the fetal period, prepuberty, puberty, or in the adult, but also to diagnose the mechanisms underlying spermatogenic dysfunction. AREAS COVERED: In this review, we principally enumerated 5 categories of testicular Sertoli cell markers - including immature Sertoli cell markers, mature Sertoli cell markers, immature/mature Sertoli cell markers, Sertoli cell functional markers, and others. EXPERT OPINION: By delineating the characteristics and applications of more than 20 Sertoli cell markers, this review provided novel Sertoli cell markers for the more accurate diagnosis and mechanistic evaluation of male reproductive disorders.


Asunto(s)
Células de Sertoli , Testículo , Adulto , Biomarcadores , Humanos , Masculino , Células de Sertoli/fisiología , Espermatogénesis , Testículo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA