Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.997
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Korean Med Sci ; 39(38): e259, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39376189

RESUMEN

BACKGROUND: While polystyrene microplastics (PS-MPs) are emerging as potentially significant health threats, linked to cancer and reproductive dysfunction, their precise effects on human health remain largely unknown. We aimed to investigate the underlying mechanisms promoting microplastic-induced damage in the reproductive system. METHODS: Thirty C57BL/6 male mice were randomly allocated into six equal-sized groups. Mice were exposed to fluorescent PS-MPs (5 µm, < 18%, green) at a dose of 1 and 3 mg/dL via oral gavage for 28 and 56 days, respectively (control, 0 mg/dL). The presence of antibodies and inflammatory and oxidative stress markers were evaluated using western blotting. Sperm analysis was also performed. Mouse testis Sertoli TM4 cells were divided into two groups: control (medium only) and PS-MPs (medium containing, 1,000 µg/mL) groups and cultured in vitro for 1, 24, 48, or 72 hours. The cells were cultured in a Ham's F12: Dulbecco's Modified Eagle Medium medium with 0.25% fetal bovine serum at 37°C with humidified atmosphere of 5% carbon dioxide in the air. Protein analyses for interleukin (IL)-6, IL-10, NADPH-oxidase (NOX)-2, NOX-4, hypoxia-inducible transcription factor (HIF)-2α, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-ß were performed using western blotting. RESULTS: The testes were evaluated after 28 and 56 days of exposure. Varying sizes of PS-MPs were detected in the testes (ranging from 5.870 to 7.768 µm). Significant differences in sperm concentration, motility, and the proportion of normal sperm were observed between the two groups. An increase in TGF-ß, HIF-2α, and NOX-4 levels was observed using western blot analysis. However, no dose-dependent correlations were observed between the two groups. In vitro evaluation of the PS-MPs group displayed PS-MP penetration of the lumen of Sertoli cells after 1 hour. Further PS-MP aggregation within Sertoli cells was observed at 24, 48, and 72 hours. A significant increase in inflammatory protein expressions (IL-10, TGF-ß, MCP-1, IL-6, TNF-α, and HIF-2α) was observed through western blotting, although oxidative agents did not show a significant increase. CONCLUSION: PS-MPs induced reproductive dysfunction in male mice provide new insights into PS-MPs-associated toxicity in mammals.


Asunto(s)
Ratones Endogámicos C57BL , Microplásticos , Estrés Oxidativo , Poliestirenos , Células de Sertoli , Masculino , Células de Sertoli/metabolismo , Células de Sertoli/efectos de los fármacos , Animales , Microplásticos/toxicidad , Microplásticos/efectos adversos , Poliestirenos/química , Poliestirenos/efectos adversos , Ratones , Estrés Oxidativo/efectos de los fármacos , Fertilidad/efectos de los fármacos , Interleucina-6/metabolismo , Motilidad Espermática/efectos de los fármacos , Testículo/metabolismo , Testículo/efectos de los fármacos , Testículo/patología , Testículo/citología , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Interleucina-10/metabolismo , Quimiocina CCL2/metabolismo , Células Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
2.
Reprod Domest Anim ; 59(10): e14729, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39387247

RESUMEN

Spermatogonial stem cells (SSCs) maintain spermatogenesis through self-renewal and differentiation. The proliferation of SSCs in culture systems can provide a valuable source of germ cells. Several studies have investigated new reproductive technologies, including the production of transgenic animals and recombinant proteins secreted from milk in goats. While studies in other species exist, research on goat SSC culture remains limited. We investigated the impact of different testosterone concentrations on the survival and colonisation of cocultured goat SSCs with Sertoli cells. Cells were isolated from immature goats using two-step enzymatic digestion and enriched by differential exclusion method. DMEM/F12 culture medium containing 1% antibiotic and 5% FBS, supplemented with GDNF (20 ng/mL), EGF, bFGF and LIF (10 ng/mL), was used with different testosterone concentrations (0, 60, 120 and 240 µg/mL) and cultured for 10 days. SC subpopulations were confirmed using PGP9.5 immunocytochemistry, and the expression of germ cell markers (ID-4, UCHL-1, THY-1, ß1-integrin, BCL6B, VASA, PLZF and OCT-4) was evaluated through RT-PCR. Alkaline phosphatase activity provided additional SSC presence. The survival rate of SSCs after isolation and the number and area of colonies on Days 4, 7 and 10 were measured using an inverted microscope. The presence of PGP 9.5 antigens and germ cell markers (ID-4, UCHL-1, THY-1, ß1-integrin, BCL6B, VASA, PLZF and OCT-4) was confirmed by immunocytochemistry and RT-PCR, respectively. According to the results, the group with 60 µg/mL testosterone had the highest number and area of colonies. The number of colonies in the 60 µg/mL testosterone group was significantly higher than the control group (p < 0.05), but no significant difference was observed compared to other groups (p ≥ 0.05). This study suggests that a low testosterone concentration (60 µg/mL) is optimal for goat SSC colonisation and viability in coculture with Sertoli cells, potentially leading to advancements in goat reproductive technologies.


Asunto(s)
Células Madre Germinales Adultas , Supervivencia Celular , Técnicas de Cocultivo , Cabras , Células de Sertoli , Testosterona , Animales , Masculino , Testosterona/farmacología , Técnicas de Cocultivo/veterinaria , Células de Sertoli/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Células Cultivadas
3.
Cell Commun Signal ; 22(1): 498, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39407201

RESUMEN

BACKGROUND: Androgens are essential hormones for testicular development and the maintenance of male fertility. Environmental factors, stress, aging, and psychological conditions can disrupt androgen production, impacting the androgen signaling pathway and consequently spermatogenesis. Within the testes, testosterone is produced by Leydig cells and acts on Sertoli cells by activating the androgen receptor (AR), which then translocates to the nucleus to function as a transcription factor. Despite clinical correlations between low testosterone levels and diminished sperm quality, the precise mechanism remains unclear. METHODS: This study explores the hypothesis that reduced androgen levels impair Sertoli cell function by disrupting AR transcriptional regulation. Using an androgen blockade model with enzalutamide, we investigated the impact of low androgen levels on AR target genes in Sertoli cells through ChIP-seq and RNA-seq assays. RESULTS: Our results reveal that androgen blockage increases AR enrichment on the promoter region of Wee1, promoting Wee1 expression, while decreasing binding to the promoter region of Lfng, inhibiting its expression. Increased WEE1 protein inhibits Sertoli cell proliferation, whereas reduced LFNG affects Notch modification, leading to decreased production of glial cell line-derived neurotrophic factor (GDNF), a key growth factor for spermatogonial stem cell self-renewal. CONCLUSIONS: These findings provide new insights into the molecular mechanisms by which low androgen levels interfere with Sertoli cell functions, offering novel perspectives for the clinical treatment of male reproductive disorders.


Asunto(s)
Andrógenos , Proliferación Celular , Nitrilos , Proteínas Tirosina Quinasas , Células de Sertoli , Animales , Masculino , Ratones , Andrógenos/farmacología , Benzamidas/farmacología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Células de Sertoli/metabolismo , Células de Sertoli/efectos de los fármacos , Células de Sertoli/citología
4.
Syst Biol Reprod Med ; 70(1): 289-298, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39361820

RESUMEN

Cryptorchidism, a condition where the testis fails to fully descend into the scrotum during development, is associated with elevated environmental temperatures and pressures, leading to male infertility and germ cell tumors. Factors such as oxidative stress and high temperatures contribute to infertility in cryptorchidism. This study aims to explore how external pressure affects Sertoli cells and discover new mechanisms affecting spermatogenesis in cryptorchidism. Sertoli cells were subjected to various pressure levels (0 mmHg, 25 mmHg, 50 mmHg, 100 mmHg) and durations (0 h, 2 h, 4 h) using an enzyme-linked immunosorbent assay (ELISA) to measure androgen binding protein (ABP) and inhibin B (INH B) secretion. Cell morphology changes were observed using immunofluorescence; apoptosis rates were measured with terminal-deoxynucleotidyl transferase mediated nick end labelling (TUNEL) assay and flow cytometry; ultrastructural variations were examined via transmission electron microscopy; and the expression of apoptosis-related proteins (Fas, FasL, caspase 3, and caspase 8) was analyzed through immunohistochemistry, real-time polymerase chain reaction (real-time PCR), and western blotting. The results showed that elevated pressure suppressed ABP and INH B secretion from Sertoli cells. Structural changes were observed under pressure, including cytoskeleton loosening and nuclear fragmentation. Apoptosis rates increased with higher pressure levels. Ultrastructural analysis revealed chromatin changes, apoptotic bodies, and mitochondrial alterations. Increased expressions of Fas and FasL were detected, along with elevated levels of caspase 3 and caspase 8. The caspase 8 inhibitor blocked pressure-induced apoptosis and caspase 3 activation, while the cytochrome C inhibitor did not show the same effect. Our findings suggested that external pressure induces apoptosis of Sertoli cells via the Fas/FasL signaling pathway, potentially contributing to male infertility associated with cryptorchidism.


Asunto(s)
Apoptosis , Proteína Ligando Fas , Células de Sertoli , Transducción de Señal , Receptor fas , Masculino , Células de Sertoli/metabolismo , Proteína Ligando Fas/metabolismo , Animales , Receptor fas/metabolismo , Presión , Ratas Sprague-Dawley , Ratas , Inhibinas/metabolismo , Espermatogénesis , Criptorquidismo/patología , Criptorquidismo/metabolismo , Células Cultivadas
5.
Ecotoxicol Environ Saf ; 285: 117114, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39357374

RESUMEN

Cadmium (Cd) has garnered significant attention due to reproductive toxicity in inducing ferroptosis. However, the specific mechanisms underlying Cd-induced germ cell ferroptosis remain poorly understood. This study aimed to systematically explore the molecular mechanisms of germ cell ferroptosis by investigating differential changes in transcription factors and proteins in male mice treated orally with CdCl2 (0.5 g/L) reaching postnatal day 60, alongside Leydig cell (TM3) and Sertoli cell (TM4) lines. Results demonstrated that Cd exposure led to increased iron overload and oxidative stress in mouse testes, disrupted intracellular mitochondrial morphology characteristic of ferroptosis. RNA sequencing revealed significant upregulation of Atf3 and Hmox1 in Cd-exposed germ cells, along with increased expression of ATF3 and HO-1. Intervention in ferroptosis or HO-1 effectively rescued cells from Cd-induced mortality by breaking the detrimental cycle between lipid peroxidation and HO-1 activation. Further findings showed that NRF2 and HO-1 expression was notably elevated upon ATF3 overexpression in TM3 and TM4 cells, activating the Keap1-Nrf2 pathway and triggering ferroptosis in testes, whereas NRF2 and HO-1 expression levels were reversed when ATF3 was silenced. This study provides novel insights into ATF3-mediated NRF2/HO-1 signaling in Cd-induced mitochondrial ferroptosis in testes, shedding light on the mechanisms underlying Cd-induced ferroptosis and testicular injury.


Asunto(s)
Factor de Transcripción Activador 3 , Cadmio , Ferroptosis , Hemo-Oxigenasa 1 , Factor 2 Relacionado con NF-E2 , Ferroptosis/efectos de los fármacos , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Masculino , Ratones , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Cadmio/toxicidad , Estrés Oxidativo/efectos de los fármacos , Testículo/efectos de los fármacos , Testículo/patología , Testículo/metabolismo , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/patología , Células Intersticiales del Testículo/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Células de Sertoli/patología , Proteínas de la Membrana
6.
Sci Rep ; 14(1): 24029, 2024 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-39402149

RESUMEN

Excess exogenous supplementation of D-galactose (D-gal), a monosaccharide and reducing sugar, generates reactive oxygen species (ROS), leading to cell damage and death. ROS accumulation is critical in aging. Therefore, D-gal-induced aging mouse models are used in aging studies. Herein, we evaluated D-gal's effect on neonatal testis development using an in vitro organ culture method. Mouse testicular fragments (MTFs) derived from neonatal testes (postnatal day 5) were cultured with 500 mM D-gal for 5 days. D-gal-treated MTFs showed a significantly increased and decreased expression of undifferentiated and differentiated germ cell markers, respectively, with a substantial reduction in meiotic cells. In D-gal-exposed MTFs, expression levels of Sertoli cell markers (Sox9 and Wt1) increased, while those of StAR and 17ß-HSD3, whose expressions are abundant in D-Gal treated adult Leydig cells, decreased. Additionally, the enzyme 3 ß-HSD1, essential for steroidogenesis in Leydig cells, was significantly reduced in D-gal-exposed MTFs compared to that in controls.D-gal significantly increased the expression of Bad, Bax, and cleaved caspase-3 and -8. Via oxidative stress in MTF. Overall, D-gal negatively regulates germ cell and Leydig cell development in neonatal testes through pro-apoptotic mechanisms and ROS.


Asunto(s)
Animales Recién Nacidos , Galactosa , Células Intersticiales del Testículo , Técnicas de Cultivo de Órganos , Testículo , Animales , Masculino , Galactosa/metabolismo , Galactosa/farmacología , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/crecimiento & desarrollo , Ratones , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/efectos de los fármacos , Técnicas de Cultivo de Órganos/métodos , Especies Reactivas de Oxígeno/metabolismo , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos
7.
Cell Rep Med ; 5(10): 101783, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39383870

RESUMEN

Sperm production depends on proper Sertoli-germ cell interaction, and we hypothesized that receptor activator of nuclear factor κB ligand (RANKL) activity in Sertoli cells may influence spermatogenesis. Treatment with the RANKL inhibitor denosumab, normally used to treat osteoporosis, increased testicular weight, inhibin B, and germ cell proliferation in ex vivo testis cultures and in vivo in a humanized RANKL mouse. The effect on germ cell proliferation was positively associated with baseline serum concentrations of anti-müllerian hormone (AMH). In accordance, denosumab increased germ cell proliferation in ex vivo human testis cultures with low/moderate but not severe impairment of Sertoli cell function. In a placebo-controlled randomized clinical trial, denosumab had no effect on semen quality but increased sperm concentration in a subgroup of infertile men with serum AMH ≥38 pmol/L at baseline. In conclusion, high serum AMH may increase the probability of a beneficial response to denosumab treatment in infertile men, thus suggesting a possible venue for precision medicine in male infertility.


Asunto(s)
Denosumab , Infertilidad Masculina , Ligando RANK , Células de Sertoli , Espermatogénesis , Masculino , Humanos , Denosumab/farmacología , Denosumab/uso terapéutico , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Espermatogénesis/efectos de los fármacos , Infertilidad Masculina/tratamiento farmacológico , Animales , Ligando RANK/metabolismo , Ratones , Hormona Antimülleriana/sangre , Proliferación Celular/efectos de los fármacos , Adulto , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología , Inhibinas/sangre , Inhibinas/metabolismo
8.
Sci Rep ; 14(1): 25965, 2024 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-39472682

RESUMEN

Numerous variables that regulate the metabolism of Sertoli cells and sperm have been identified, one of which is sex steroid hormones. These hormones play a vital role in maintaining energy homeostasis, influencing the overall metabolic balance of the human body. The proper functioning of the reproductive system is closely linked to energy status, as the reproductive axis responds to metabolic signals. The aim of this study was to investigate the gene expression patterns of metabolite interconversion enzymes in testicular cells (Sertoli cells and spermatogonia) of non-obstructive azoospermia (NOA) patients, as compared to normal controls, to understand the molecular mechanisms contributing to NOA. We used microarray and bioinformatics techniques to analyze 2912 genes encoding metabolite interconversion enzymes, including methyltransferase, monooxygenase, transmembrane reductase, and phosphohydrolase, in both testicular cells and normal samples. In sperm, the upregulation of MOXD1, ACAD10, PCYT1A, ARG1, METTL6, GPLD1, MAOA, and CYP46A1 was observed, while ENTPD2, CPT1C, ADC, and CYB5B were downregulated. Similarly, in the Sertoli cells of three NOA patients, RPIA, PIK3C3, LYPLA2, CA11, MBOAT7, and HDHD2 were upregulated, while NAA25, MAN2A1, CYB561, PNPLA5, RRM2, and other genes were downregulated. Using STRING and Cytoscape, we predicted the functional and molecular interactions of these proteins and identified key hub genes. Pathway enrichment analysis highlighted significant roles for G1/S-specific transcription, pyruvate metabolism, and citric acid metabolism in sperm, and the p53 signaling pathway and folate metabolism in Sertoli cells. Additionally, Weighted Gene Co-expression Network Analysis (WGCNA) and single-cell RNA sequencing (scRNA-seq) were performed to validate these findings, revealing significant alterations in gene expression and cellular distribution in NOA patients. Together, these results provide new insights into the molecular mechanisms underlying NOA and identify potential therapeutic targets.


Asunto(s)
Azoospermia , Células de Sertoli , Espermatozoides , Masculino , Humanos , Azoospermia/genética , Azoospermia/metabolismo , Azoospermia/patología , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Simulación por Computador , Biología Computacional/métodos , Perfilación de la Expresión Génica , Adulto
9.
Pestic Biochem Physiol ; 205: 106175, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39477627

RESUMEN

Resmethrin, a pyrethroid pesticide used to control insects, is toxic to non-target organisms and other mammals. However, little is known about the reproductive toxicity of resmethrin in the testes, or its mechanism of toxicity. In this study, we investigated the testicular toxicity of resmethrin on mouse Leydig (TM3) and Sertoli (TM4) cells, focusing on the mitochondria and endoplasmic reticulum (ER). We found that resmethrin inhibited proliferation and cell cycle progression and disrupted mitochondrial membrane potential (MMP; ΔΨ) in TM3 and TM4 cells. In particular, resmethrin exposure significantly reduced the expression of mitochondria-associated membranes (MAMs) proteins, such as Vapb, Vdac, and Grp75, in both cell lines. Resmethrin also disrupts calcium homeostasis in the mitochondrial matrix and cytoplasm. In addition, resmethrin activates oxidative stress-mediated ER stress signals. Finally, we confirmed that 4-PBA, an ER stress inhibitor, restored the growth of TM3 and TM4 cells, which was decreased by resmethrin. Therefore, we confirmed that resmethrin hampered MAMs and activated ER stress, thus suppressing TM3 and TM4 cell proliferation.


Asunto(s)
Proliferación Celular , Estrés del Retículo Endoplásmico , Células Intersticiales del Testículo , Potencial de la Membrana Mitocondrial , Piretrinas , Células de Sertoli , Animales , Ratones , Masculino , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Piretrinas/toxicidad , Piretrinas/farmacología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Insecticidas/toxicidad , Insecticidas/farmacología , Membranas Asociadas a Mitocondrias , Butilaminas
10.
Cell Reprogram ; 26(5): 135-145, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39475771

RESUMEN

Spermatogenesis constitutes a complex and intricate cascade of differentiation, indispensable for the male reproductive competence. The intercellular communication conduits of Sertoli cells (SCs) are pivotal in orchestrating this cascade ensuring sustenance and development of germ cells. Single cells and bioinformatics recently demonstrated articles are used for the regulatory modalities through which SCs modulate spermatogenesis, specifically via androgen receptors (ARs), the transforming growth factor-beta/Smad axis, mitogen-activated protein kinases, cAMP/protein kinase A (PKA), phosphatidylinositol 4,5-bisphosphate 3-kinase (PI3k)/AKT serine threonine kinase (Akt), AMP-activated protein kinase, and AR pathways. Within this framework, homeostasis of gap junction dynamics, cryptic sites and the activities at tight junctions and adherens junctions, with the integrity of the testicular barrier, glucose assimilation, lactate distribution, being governed also along with SC maturation. Disruptions in activities or abnormal concentration in derangements in AR, cAMP/PKA, and PI3k/Akt pathways, and as well as the molecules that comprise them, would present male infertility.


Asunto(s)
Células de Sertoli , Transducción de Señal , Análisis de la Célula Individual , Células de Sertoli/metabolismo , Células de Sertoli/citología , Masculino , Humanos , Animales , Ontología de Genes , Espermatogénesis
11.
Genes (Basel) ; 15(9)2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39336792

RESUMEN

Blood-testis barrier (BTB) genes are crucial for the cellular mechanisms of spermatogenesis as they protect against detrimental cytotoxic agents, chemicals, and pathogens, thereby maintaining a sterile environment necessary for sperm development. BTB proteins predominantly consist of extensive tight and gap junctions formed between Sertoli cells. These junctions form a crucial immunological barrier restricting the intercellular movement of substances and molecules within the adluminal compartment. Epithelial tight junctions are complex membrane structures composed of various integral membrane proteins, including claudins, zonula occludens-1, and occludin. Inter-testicular cell junction proteins undergo a constant process of degradation and renewal. In addition, the downregulation of genes crucial to the development and preservation of cell junctions could disrupt the functionality of the BTB, potentially leading to male infertility. Oxidative stress and inflammation may contribute to disrupted spermatogenesis, resulting in male infertility. L-cysteine is a precursor to glutathione, a crucial antioxidant that helps mitigate damage and inflammation resulting from oxidative stress. Preclinical research indicates that L-cysteine may offer protective benefits against testicular injury and promote the expression of BTB genes. This review emphasizes various BTB genes essential for preserving its structural integrity and facilitating spermatogenesis and male fertility. Furthermore, it consolidates various research findings suggesting that L-cysteine may promote the expression of BTB-associated genes, thereby aiding in the maintenance of testicular functions.


Asunto(s)
Barrera Hematotesticular , Cisteína , Espermatogénesis , Masculino , Barrera Hematotesticular/metabolismo , Humanos , Animales , Cisteína/metabolismo , Uniones Estrechas/metabolismo , Estrés Oxidativo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/efectos de los fármacos , Testículo/metabolismo
12.
Stem Cell Res Ther ; 15(1): 282, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39227878

RESUMEN

BACKGROUND: Inflammation-induced testicular damage is a significant contributing factor to the increasing incidence of infertility. Traditional treatments during the inflammatory phase often fail to achieve the desired fertility outcomes, necessitating innovative interventions such as cell therapy. METHODS: We explored the in vivo properties of intravenously administered Sertoli cells (SCs) in an acute lipopolysaccharide (LPS)-induced inflammatory mouse model. Infiltrating and resident myeloid cell phenotypes were assessed using flow cytometry. The impact of SC administration on testis morphology and germ cell quality was evaluated using computer-assisted sperm analysis (CASA) and immunohistochemistry. RESULTS: SCs demonstrated a distinctive migration pattern, importantly they preferentially concentrated in the testes and liver. SC application significantly reduced neutrophil infiltration as well as preserved the resident macrophage subpopulations. SCs upregulated MerTK expression in both interstitial and peritubular macrophages. Applied SC treatment exhibited protective effects on sperm including their motility and kinematic parameters, and maintained the physiological testicular morphology. CONCLUSION: Our study provides compelling evidence of the therapeutic efficacy of SC transplantation in alleviating acute inflammation-induced testicular damage. These findings contribute to the expanding knowledge on the potential applications of cell-based therapies for addressing reproductive health challenges and offer a promising approach for targeted interventions in male infertility.


Asunto(s)
Inflamación , Células de Sertoli , Espermatozoides , Masculino , Animales , Células de Sertoli/metabolismo , Ratones , Inflamación/patología , Inflamación/terapia , Espermatozoides/metabolismo , Lipopolisacáridos/toxicidad , Ratones Endogámicos C57BL , Testículo , Tirosina Quinasa c-Mer/metabolismo , Tirosina Quinasa c-Mer/genética , Motilidad Espermática , Macrófagos/metabolismo
13.
Pestic Biochem Physiol ; 204: 106065, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39277380

RESUMEN

Organophosphate pesticides have potent endocrine disrupting effects, hence banned in many countries. However, many organophosphates like chlorpyrifos, malathion et cetera continue to be used in some countries (Wolejko et al., 2022; Wolejko et al., 2022)including India. Fodder mediated ingestion of these substances may be harmful for livestock fertility. We have investigated the effect of the widely used organophosphate pesticide chlorpyrifos (CPF) and its metabolite, 3,5,6-trichloropyridinol (TCPy) on the expression of genes essential for spermatogenesis in goat testicular tissue. The testicular Sertoli cells (Sc) regulate germ cell division and differentiation under the influence of follicle stimulating hormone (FSH) and testosterone (T). Impaired FSH and T mediated signalling in Sc can compromise spermatogenesis leading to sub-fertility/infertility. As Sc express receptors (R) for FSH and T, they are highly susceptible to the endocrine disrupting effects of pesticides affecting fertility by dysregulating the functioning of Sc. Our results indicated that exposure to different concentrations of CPF and TCPy can compromise Sc function by downregulating the expression of FSHR and AR which was associated with a concomitant decline in the expression of genes essential for germ cell division and differentiation, like KITLG, INHBB, CLDN11 and GJA1. CPF also induced a significant reduction in the activity of acetylcholinesterase in the testes and increased the total testicular antioxidant capacity. Our results suggested that CPF and its metabolite TCPy may induce reproductive toxicity by dysregulating the expression of Sc specific genes essential for spermatogenesis.


Asunto(s)
Cloropirifos , Cabras , Espermatogénesis , Testículo , Animales , Masculino , Espermatogénesis/efectos de los fármacos , Cloropirifos/toxicidad , Testículo/efectos de los fármacos , Testículo/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Insecticidas/toxicidad , Piridinas/farmacología , Piridinas/toxicidad , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Piridonas
14.
Mol Biol Rep ; 51(1): 982, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39271559

RESUMEN

BACKGROUND: The Axl gene is a receptor tyrosine kinase essential for male fertility. With other Tyro3 family members, it regulates cell apoptosis and preserves the organization of seminiferous tubules. However, the regulation of the expression of Axl in testicular Sertoli cells is not entirely understood. The transcription factors NR5A1 and JUNB are involved in several male fertility mechanisms such as sex development and steroidogenesis. We hypothesize that Axl promoter activity is regulated by cooperation between JUNB and NR5A1 in Sertoli cells. METHODS AND RESULTS: Following transfections of TM4 Sertoli cells with DsiRNA interference against Axl, our results show that cell morphology may be regulated by AXL. Using transfections of expression plasmids and reporter plasmids containing the Axl promoter, we report that Axl expression is highly activated by cooperation between NR5A1 and JUNB in TM4 and 15P-1 Sertoli cells. Chromatin immunoprecipitation and luciferase reporter assays with 5' promoter deletions demonstrate that JUNB and NR5A1 are being recruited to DNA regulatory elements in the proximal region of the Axl promoter. The fourth intronic region of Axl also participates in the recruitment of JUNB. CONCLUSION: Thus, Axl expression is regulated by a cooperation between the transcription factors JUNB and NR5A1 and influences the morphology of TM4 Sertoli cells.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Células de Sertoli , Factor Esteroidogénico 1 , Factores de Transcripción , Animales , Células de Sertoli/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica
15.
Elife ; 132024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39283662

RESUMEN

Over the past several decades, a trend toward delayed childbirth has led to increases in parental age at the time of conception. Sperm epigenome undergoes age-dependent changes increasing risks of adverse conditions in offspring conceived by fathers of advanced age. The mechanism(s) linking paternal age with epigenetic changes in sperm remain unknown. The sperm epigenome is shaped in a compartment protected by the blood-testes barrier (BTB) known to deteriorate with age. Permeability of the BTB is regulated by the balance of two mTOR complexes in Sertoli cells where mTOR complex 1 (mTORC1) promotes the opening of the BTB and mTOR complex 2 (mTORC2) promotes its integrity. We hypothesized that this balance is also responsible for age-dependent changes in the sperm epigenome. To test this hypothesis, we analyzed reproductive outcomes, including sperm DNA methylation in transgenic mice with Sertoli cell-specific suppression of mTORC1 (Rptor KO) or mTORC2 (Rictor KO). mTORC2 suppression accelerated aging of the sperm DNA methylome and resulted in a reproductive phenotype concordant with older age, including decreased testes weight and sperm counts, and increased percent of morphologically abnormal spermatozoa and mitochondrial DNA copy number. Suppression of mTORC1 resulted in the shift of DNA methylome in sperm opposite to the shift associated with physiological aging - sperm DNA methylome rejuvenation and mild changes in sperm parameters. These results demonstrate for the first time that the balance of mTOR complexes in Sertoli cells regulates the rate of sperm epigenetic aging. Thus, mTOR pathway in Sertoli cells may be used as a novel target of therapeutic interventions to rejuvenate the sperm epigenome in advanced-age fathers.


Asunto(s)
Metilación de ADN , Células de Sertoli , Espermatozoides , Masculino , Animales , Células de Sertoli/metabolismo , Ratones , Espermatozoides/metabolismo , Espermatozoides/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Serina-Treonina Quinasas TOR/metabolismo , Ratones Noqueados , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Proteína Reguladora Asociada a mTOR/metabolismo , Proteína Reguladora Asociada a mTOR/genética , Ratones Transgénicos , Envejecimiento/fisiología , Envejecimiento/genética , Transducción de Señal , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Epigénesis Genética
16.
J Endocrinol ; 263(3)2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39303739

RESUMEN

Liraglutide, an analog of the incretin hormone glucagon-like peptide 1 (GLP-1), is widely used for obesity and type 2 diabetes treatment. However, there is scarce information about its effects on testicular function. Within the testis, Sertoli cells (SCs) provide nutritional support for germ cells; they metabolize glucose to lactate, which is delivered to germ cells to be used as a preferred energy substrate. Besides, SCs use fatty acids (FAs) as an energy source and store them as triacylglycerols (TAGs) within lipid droplets (LDs), which serve as an important energy reserve. In the present study, 20-day-old rat SC cultures were used to assess whether liraglutide affects their metabolic functions related to nutritional support and lipid storage. The results show that liraglutide does not modify glucose consumption or lactate production. However, it increases TAG levels and LD content. These effects are accompanied by an increase in the mRNA levels of the fatty acid transporter FAT/CD36, glycerol-3-phosphate-acyltransferase 3, and perilipins 1 and 4. The participation of the cAMP/PKA signaling pathway was explored. We observed that H89 (a PKA inhibitor) decreases the LD upregulation elicited by liraglutide, and that dibutyryl cAMP increases LD content and the expression of related genes. In summary, liraglutide promotes lipid storage in SCs through the regulation of key regulatory genes involved in FA transport, TAG synthesis, and LD formation. Considering the importance of lipid storage in SC energetic homeostasis maintenance, we postulate that liraglutide might improve the overall energetic status of the seminiferous tubule.


Asunto(s)
Metabolismo Energético , Liraglutida , Células de Sertoli , Animales , Masculino , Liraglutida/farmacología , Células de Sertoli/metabolismo , Células de Sertoli/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ratas , Células Cultivadas , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/farmacología , Triglicéridos/metabolismo , Glucosa/metabolismo , Gotas Lipídicas/metabolismo , Gotas Lipídicas/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Antígenos CD36/metabolismo , Antígenos CD36/genética
17.
FASEB J ; 38(18): e70049, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39275889

RESUMEN

In this study, we have explored the role of the KATNB1 gene, a microtubule-severing protein, in the seminiferous epithelium of the rat testis. Our data have shown that KATNB1 expressed in rat brain, testes, and Sertoli cells. KATNB1 was found to co-localize with α-tubulin showing a unique stage-specific distribution across the seminiferous epithelium. Knockdown of KATNB1 by RNAi led to significant disruption of the tight junction (TJ) permeability barrier function in primary Sertoli cells cultured in vitro with an established functional TJ-barrier, as well as perturbations in the microtubule and actin cytoskeleton organization. The disruption in these cytoskeletal structures, in turn, led to improper distribution of TJ and basal ES proteins essential for maintaining the Sertoli TJ function. More importantly, overexpression of KATNB1 in the testis in vivo was found to block cadmium-induced blood-testis barrier (BTB) disruption and testis injury. KATNB1 exerted its promoting effects on BTB and spermatogenesis through corrective spatiotemporal expression of actin- and microtubule-based regulatory proteins by maintaining the proper organization of cytoskeletons in the testis, illustrating its plausible therapeutic implication. In summary, Katanin regulatory subunit B1 (KATNB1) plays a crucial role in BTB and spermatogenesis through its effects on the actin- and microtubule-based cytoskeletons in Sertoli cells and testis, providing important insights into male reproductive biology.


Asunto(s)
Barrera Hematotesticular , Katanina , Células de Sertoli , Animales , Masculino , Células de Sertoli/metabolismo , Ratas , Katanina/metabolismo , Katanina/genética , Barrera Hematotesticular/metabolismo , Citoesqueleto/metabolismo , Ratas Sprague-Dawley , Uniones Estrechas/metabolismo , Espermatogénesis/fisiología , Células Cultivadas , Epitelio Seminífero/metabolismo , Testículo/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo
18.
Ecotoxicol Environ Saf ; 284: 116880, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39142115

RESUMEN

Past studies have observed that BHPF induces multi-organ toxicity, however, whether it induces damage to male reproductive system and the specific mechanism remains unclear. In the present study, male mice were given 0, 2, 10 or 50 mg/kg/day of BHPF by gavage for 35 days to observe its effect on reproductive organ and sperm quality. The results indicated that BHPF decreased sperm count and sperm motility in a dose-dependent manner. Besides, our results demonstrated that BHPF triggered the proliferation inhibition and cell death of germ cells in vivo and in vitro. Also, BHPF reduced the expression of function markers for germ cells, Sertoli cells, and Leydig cells, indicating its damage to function of testis cells. Simultaneously, testicular microenvironment was found to be altered by BHPF, as presented with declined testosterone level and decreased expression of local microenvironment regulators. Overall, our findings indicated the detrimental effects of BHPF on male reproductive function in mice, suggesting testicular function and local microenvironment disturbance as mechanism underlying testicular damage.


Asunto(s)
Fertilidad , Recuento de Espermatozoides , Motilidad Espermática , Testículo , Masculino , Animales , Testículo/efectos de los fármacos , Testículo/patología , Ratones , Motilidad Espermática/efectos de los fármacos , Fertilidad/efectos de los fármacos , Testosterona/sangre , Células Intersticiales del Testículo/efectos de los fármacos , Células de Sertoli/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Compuestos de Bencidrilo/toxicidad , Fenoles/toxicidad , Proliferación Celular/efectos de los fármacos
19.
Hum Reprod ; 39(10): 2210-2220, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39198007

RESUMEN

STUDY QUESTION: Do testis-specific cells have a normal karyotype in non-mosaic postpubertal Klinefelter syndrome (KS) patients with focal spermatogenesis and in non-mosaic prepubertal KS boys? SUMMARY ANSWER: Spermatogonia have a 46, XY karyotype, and Sertoli cells surrounding these spermatogonia in postpubertal patients also have a 46, XY karyotype, whereas, in prepubertal KS boys, Sertoli cells surrounding the spermatogonia still have a 47, XXY karyotype. WHAT IS KNOWN ALREADY: A significant proportion of patients with non-mosaic KS can have children by using assisted reproductive techniques thanks to focal spermatogenesis. However, the karyotype of the cells that are able to support focal spermatogenesis has not been revealed. STUDY DESIGN, SIZE, DURATION: Testicular biopsy samples from non-mosaic KS patients were included in the study. Karyotyping for sex chromosomes in testis-specific cells was performed by immunohistochemical analysis of inactive X (Xi) chromosome and/or fluorescent in situ hybridization (FISH) analysis of chromosomes 18, X, and Y. PARTICIPANTS/MATERIALS, SETTING, METHODS: A total of 22 KS patients (17 postpubertal and 5 prepubertal) who were non-mosaic according to lymphocyte karyotype analysis, were included in the study. After tissue processing, paraffin embedding, and sectioning, the following primary antibodies were used for cell-specific analysis and Xi detection; one section was stained with MAGE A4 for spermatogonia, SOX9 for Sertoli cells, and H3K27me3 for Xi; the other one was stained with CYP17A1 for Leydig cells, ACTA2 for peritubular myoid cells, and H3K27me3 for Xi. Xi negative (Xi-) somatic cells (i.e. Sertoli cells, Leydig cells, and peritubular myoid cells) were evaluated as having the 46, XY karyotype; Xi positive (Xi+) somatic cells were evaluated as having the 47, XXY. FISH stain for chromosomes 18, X, and Y was performed on the same sections to investigate the karyotype of spermatogonia and to validate the immunohistochemistry results for somatic cells. MAIN RESULTS AND THE ROLE OF CHANCE: According to our data, all spermatogonia in both postpubertal and prepubertal non-mosaic KS patients seem to have 46, XY karyotype. However, while the Sertoli cells surrounding spermatogonia in postpubertal samples also had a 46, XY karyotype, the Sertoli cells surrounding spermatogonia in prepubertal samples had a 47, XXY karyotype. In addition, while the Sertoli cells in some of the Sertoli cell-only tubules had 46, XY karyotype, the Sertoli cells in some of the other Sertoli cell-only tubules had 47, XXY karyotype in postpubertal samples. In contrast to the postpubertal samples, Sertoli cells in all tubules in the prepubertal samples had the 47, XXY karyotype. Our data also suggest that germ cells lose the extra X chromosome during embryonic, fetal, or neonatal life, while Sertoli cells lose it around puberty. Peritubular myoid cells and Leydig cells may also be mosaic in both postpubertal patients and prepubertal boys, but it requires further investigation. LIMITATIONS, REASONS FOR CAUTION: The number of prepubertal testicle samples containing spermatogonia is limited, so more samples are needed for a definitive conclusion. The fact that not all the cell nuclei coincide with the section plane limits the accurate detection of X chromosomes by immunohistochemistry and FISH in some cells. To overcome this limitation, X chromosome analysis could be performed by different techniques on intact cells isolated from fresh tissue. Additionally, there is no evidence that X chromosome inactivation reoccurs after activation of the Xi during germ cell migration during embryogenesis, limiting the prediction of X chromosome content in germ cells by H3K27me3. WIDER IMPLICATIONS OF THE FINDINGS: Our findings will lay the groundwork for new clinically important studies on exactly when and by which mechanism an extra X chromosome is lost in spermatogonia and Sertoli cells. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by The Scientific and Technological Research Council of Türkiye (TUBITAK) (2219 - International Postdoctoral Research Fellowship Program for Turkish Citizens) and the Strategic Research Program (SRP89) from the Vrije Universiteit Brussel. The authors declare no competing interests. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Síndrome de Klinefelter , Mosaicismo , Células de Sertoli , Espermatogénesis , Espermatogonias , Testículo , Humanos , Masculino , Síndrome de Klinefelter/genética , Espermatogénesis/genética , Niño , Testículo/patología , Testículo/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patología , Espermatogonias/metabolismo , Adolescente , Cariotipificación , Preescolar , Pubertad , Cariotipo , Hibridación Fluorescente in Situ
20.
Reproduction ; 168(5)2024 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-39121036

RESUMEN

In brief: FSH leads to glutamine dependence, which is required for mTORC1 activation and in consequence Sertoli cell proliferation. Abstract: The spermatogenic capacity of adult individuals depends on, among other factors, the number of Sertoli cells (SCs) that result from the proliferative waves during development. FSH upregulates SC proliferation at least partly, through the activation of the PI3K/Akt/mTORC1 pathway, among other mechanisms. It is widely known that mTORC1 is a sensor of amino acids. Among amino acids, glutamine acquires relevance since it might contribute to cell cycle progression through the modulation of mTORC1 activity. It has not been studied yet whether glutamine intervenes in FSH-mediated regulation of SC proliferation and cell cycle progression, or if FSH has any effect on glutamine metabolism. Eight-day-old rat SCs were incubated in culture media without glutamine or with glutamine in the absence or presence of a glutamine transporter inhibitor or a glutaminase activity inhibitor under basal conditions or stimulated with FSH. The results obtained show that FSH does not promote SC proliferation and mTORC1 activation in the absence of glutamine. Also, FSH modulates glutamine metabolism increasing glutaminase isoform 2 and reducing glutamine synthetaseexpression. FSH did not promote SC proliferation and mTORC1 activation when glutaminase activity was inhibited. The results suggest that glutamine or its metabolites might cooperate with FSH in the upregulation of SC proliferation through mTORC1. In addition, as FSH modulates glutamine metabolism through the induction of glutaminase isoform 2, the hormonal control of glutamine metabolism might be part of the intricate signaling network triggered by FSH, which is crucial to establish the population of mature SCs that supports the reproductive function.


Asunto(s)
Proliferación Celular , Hormona Folículo Estimulante , Glutamina , Diana Mecanicista del Complejo 1 de la Rapamicina , Células de Sertoli , Animales , Glutamina/metabolismo , Glutamina/farmacología , Masculino , Células de Sertoli/metabolismo , Células de Sertoli/efectos de los fármacos , Células de Sertoli/citología , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/metabolismo , Proliferación Celular/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratas , Células Cultivadas , Transducción de Señal/efectos de los fármacos , Glutaminasa/metabolismo , Ratas Sprague-Dawley , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA