Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 778
Filter
1.
Syst Biol Reprod Med ; 70(1): 289-298, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39361820

ABSTRACT

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.


Subject(s)
Apoptosis , Fas Ligand Protein , Sertoli Cells , Signal Transduction , fas Receptor , Male , Sertoli Cells/metabolism , Fas Ligand Protein/metabolism , Animals , fas Receptor/metabolism , Pressure , Rats, Sprague-Dawley , Rats , Inhibins/metabolism , Spermatogenesis , Cryptorchidism/pathology , Cryptorchidism/metabolism , Cells, Cultured
2.
Urol Case Rep ; 57: 102854, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39391707

ABSTRACT

Sertoli cell tumors are a rare subtype of testicular tumors. This report describes a 55-year-old male who presented with scrotal pain and a palpable mass. Diagnostic imaging revealed a hypoechoic mass in the left epididymis and a hyperechoic mass in the right testis. A right testis-sparing surgical procedure was performed, and subsequent histopathological analysis confirmed the presence of a benign Sertoli cell tumor. The patient experienced an uncomplicated postoperative course and was discharged on the same day. This case underscores the viability of testis-sparing surgery in the management of rare testicular tumors, emphasizing its potential for preserving testicular function.

3.
Article in English | MEDLINE | ID: mdl-39390674

ABSTRACT

Myosin VI has been reported by others to localize in association with various regions of apical tubulobulbar complexes (TBCs) at sites of attachment between Sertoli cells and late spermatids in the mouse. Tubulobulbar complexes internalize "intact" intercellular junctions during sperm release and during spermatocyte translocation through the blood-testis barrier. Here, we use super-resolution (STED-stimulated emission depletion) and electron microscopy of immunolabeled sections of rat testis to clearly define the localization of anti-myosin VI reactivity both at apical and basal sites in the epithelium. In data stacks collected by STED imaging, staining at TBCs was predominantly associated with bulb regions of the complexes. At apical sites, when data stacks were analyzed with an Imaris software, staining appeared around and extended between adjacent bulbs. At basal sites, in addition to labeling at TBC bulbs, reactive sites appeared concentrated in regions close to but not directly associated with intercellular junctions. At the ultrastructural level, labeling was predominantly associated with cisternae of the endoplasmic reticulum associated with the bulbs of TBCs and near to basal junction complexes. We conclude that myosin VI may be associated with specific subdomains of the endoplasmic reticulum related to TBC bulbs and associated basal junction complexes between Sertoli cells.

4.
Endocrinology ; 165(11)2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39279445

ABSTRACT

Thyroid hormone regulates the rate of testis maturation in mammals. Manipulations of thyroid hormone levels in neonatal animals affect various aspects of testis biology. However, there have been no studies examining the effects of thyroid hormone on the rete testis (RT). Here, we used animal models of neonatal hyperthyroidism (injections of triiodothyronine, or T3) and hypothyroidism (goitrogen 6-propyl-2-thiouracil [PTU] treatment) and found that higher levels of thyroid hormone accelerate RT development, while lower levels of thyroid hormone delay it. T3 and PTU treatments influence RT size, proliferation of RT cells, and expression of DMRT1 and androgen receptor in the RT. T3 supplementation accelerates RT development in an organ testicular culture, which indicates the local action of thyroid hormone. Additionally, it was found that follicle-stimulating hormone could be involved in the regulation both of RT proliferation and RT size. The fact that RT cells in a cell culture do not respond to T3 suggests indirect action of thyroid hormone on the RT in vivo or the loss of the responsiveness to the hormone in vitro.


Subject(s)
Animals, Newborn , Testis , Thyroid Hormones , Triiodothyronine , Animals , Male , Mice , Testis/drug effects , Testis/metabolism , Testis/growth & development , Triiodothyronine/pharmacology , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Propylthiouracil/pharmacology , Cell Proliferation/drug effects , Hyperthyroidism/chemically induced , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Hypothyroidism/chemically induced , Follicle Stimulating Hormone/pharmacology , Receptors, Androgen/metabolism
5.
Pestic Biochem Physiol ; 204: 106065, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277380

ABSTRACT

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.


Subject(s)
Chlorpyrifos , Goats , Spermatogenesis , Testis , Animals , Male , Spermatogenesis/drug effects , Chlorpyrifos/toxicity , Testis/drug effects , Testis/metabolism , Down-Regulation/drug effects , Insecticides/toxicity , Pyridines/pharmacology , Pyridines/toxicity , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Pyridones
6.
Toxicol In Vitro ; 101: 105941, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39278285

ABSTRACT

Chloroquine (CQ) is widely used in the therapy against malarial, tumor and recently the COVID-19 pandemic, as a lysosomotropic agent to inhibit the endolysosomal trafficking in the autophagy pathway. We previously reported that CQ (20 µM, 36 h) could reprogram transcriptome, and impair multiple signaling pathways vital to porcine immature Sertoli cells (iSCs). However, whether CQ treatment could affect the metabolomic compositions of porcine iSCs remains unclear. Here, we showed that CQ (20 µM, 36 h) treatment of porcine iSCs induced significant changes of 63 metabolites (11 up and 52 down) by the metabolomics method, which were involved in different metabolic pathways. Caffeic acid and esculetin, the top two up-regulated metabolites, were validated by ELISA. The combined analysis of metabolomics and transcriptome showed caffeic acid and esculetin to be highly correlated with multiple differentially expressed genes (DEGs), including Ndrg1, S100a8, Sqstm1, S100a12, S100a9, Ill1, Lif, Ntn4 and Peg10. Furthermore, esculetin treatment (53 nM, 36 h) significantly decreased the viability and proliferation, suppressed the mitochondrial function, whereas promoted the apoptosis of porcine iSCs, similar to those by CQ treatment (20 µM, 36 h). Collectively, our results showed that CQ treatment induces metabolic changes, and its effect on porcine iSCs could be partially mediated by esculetin.

7.
FASEB J ; 38(18): e70049, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39275889

ABSTRACT

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.


Subject(s)
Blood-Testis Barrier , Katanin , Sertoli Cells , Animals , Male , Sertoli Cells/metabolism , Rats , Katanin/metabolism , Katanin/genetics , Blood-Testis Barrier/metabolism , Cytoskeleton/metabolism , Rats, Sprague-Dawley , Tight Junctions/metabolism , Spermatogenesis/physiology , Cells, Cultured , Seminiferous Epithelium/metabolism , Testis/metabolism , Microtubules/metabolism , Tubulin/metabolism
8.
J Ayurveda Integr Med ; 15(4): 100930, 2024.
Article in English | MEDLINE | ID: mdl-39121783

ABSTRACT

BACKGROUND: Medications, including chemotherapeutic drugs, contribute to male infertility as external factors by inducing oxidative stress in testicular cells. Shilajit is a naturally occurring bioactive antioxidant used in Ayurvedic medicine to treat a variety of ailments. OBJECTIVE: This study examines the potential of Shilajit to counteract the negative effects of the chemotherapeutic drug cyclophosphamide (CPA) on testicular germ cell dynamics. MATERIAL AND METHODS: Male Parkes mice received single intraperitoneal CPA injection (200 mg/kg BW) on day one, followed by daily supplementation of Shilajit (100 and 200 mg/kg BW) for one spermatogenic cycle. RESULTS: CPA adversely affected testicular germ cell dynamics by inhibiting the conversion of spermatogonia-to-spermatids, altering testicular histoarchitecture, impairing Sertoli cell function and testicular steroidogenesis, and disturbing the testicular oxido-apoptotic balance. Shilajit supplementation restores testicular germ cell dynamics in CPA-exposed mice, as evidenced by improved histoarchitecture of the testis. Shilajit improves testicular daily production and sperm quality by promoting the conversion of spermatogonia (2C) into spermatids (1C), stimulating germ cell proliferation (PCNA), improving Sertoli cell function (N-Cadherin and ß-Catenin), and maintaining the Bax/Bcl2 ratio. Additionally, Shilajit enhances testosterone biosynthesis by activating enzymes like 3ß-HSD, and 17ß-HSD. Shilajit also reduces testicular oxidative stress by increasing antioxidant enzyme activity (SOD) and decreasing lipid peroxidation (LPO). These effects are mediated by upregulation of the antioxidant protein Nrf-2 and downregulation of Keap-1. CONCLUSION: The findings underscore the potent androgenic and antioxidant characteristics of Shilajit, as well as its ability to enhance fertility in cases of testicular damage caused by chemotherapeutic drugs.

9.
Sci Rep ; 14(1): 18418, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117695

ABSTRACT

Bacterial testicular inflammation is one of the important causes of male infertility. Using plant-derived compounds to overcome the side effects of antibiotics is an alternative treatment strategy for many diseases. Schizandrin B (SchB) is a bioactive compound of herbal medicine Schisandra chinensis which has multiple pharmacological effects. However its effect and the mechanism against testicular inflammation are unknown. Here we tackled these questions using models of lipopolysaccharide (LPS)-induced mice and -Sertoli cells (SCs). Histologically, SchB ameliorated the LPS-induced damages of the seminiferous epithelium and blood-testicular barrier, and reduced the production of pro-inflammatory mediators in mouse testes. Furthermore, SchB decreased the levels of pro-inflammatory mediators and inhibited the nuclear factor kB (NF-κB) and MAPK (especially JNK) signaling pathway phosphorylation in LPS-induced mSCs. The bioinformatics analysis based on receptor prediction and the molecular docking was further conducted. We targeted androgen receptor (AR) and illustrated that AR might bind with SchB in its function. Further experiments indicate that the AR expression was upregulated by LPS stimulation, while SchB treatment reversed this phenomenon; similarly, the expression of the JNK-related proteins and apoptotic-related protein were also reversed after AR activator treatment. Together, SchB mitigates LPS-induced inflammation and apoptosis by inhibiting the AR-JNK pathway.


Subject(s)
Apoptosis , Cyclooctanes , Lignans , Lipopolysaccharides , Polycyclic Compounds , Sertoli Cells , Animals , Male , Cyclooctanes/pharmacology , Polycyclic Compounds/pharmacology , Polycyclic Compounds/therapeutic use , Lignans/pharmacology , Lignans/therapeutic use , Apoptosis/drug effects , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, Androgen/metabolism , MAP Kinase Signaling System/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Molecular Docking Simulation , Testis/drug effects , Testis/metabolism , Testis/pathology , NF-kappa B/metabolism
10.
J Pathol ; 264(3): 250-269, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39161125

ABSTRACT

Testicular tumors represent the most common malignancy among young men. Nevertheless, the pathogenesis and molecular underpinning of testicular tumors remain largely elusive. We aimed to delineate the intricate intra-tumoral heterogeneity and the network of intercellular communication within the tumor microenvironment. A total of 40,760 single-cell transcriptomes were analyzed, encompassing samples from six individuals with seminomas, two patients with mixed germ cell tumors, one patient with a Leydig cell tumor, and three healthy donors. Five distinct malignant subclusters were identified in the constructed landscape. Among them, malignant 1 and 3 subclusters were associated with a more immunosuppressive state and displayed worse disease-free survival. Further analysis identified that APP-CD74 interactions were significantly strengthened between malignant 1 and 3 subclusters and 14 types of immune subpopulations. In addition, we established an aberrant spermatogenesis trajectory and delineated the global gene alterations of somatic cells in seminoma testes. Sertoli cells were identified as the somatic cell type that differed the most from healthy donors to seminoma testes. Cellular communication between spermatogonial stem cells and Sertoli cells is disturbed in seminoma testes. Our study delineates the intra-tumoral heterogeneity and the tumor immune microenvironment in testicular tumors, offering novel insights for targeted therapy. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Testicular Neoplasms , Tumor Microenvironment , Humans , Male , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology , Testicular Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Gene Expression Profiling/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Transcriptome , Disease Progression , Gene Expression Regulation, Neoplastic , Seminoma/genetics , Seminoma/pathology , Seminoma/immunology , Immune Tolerance/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/immunology , Antigens, Differentiation, B-Lymphocyte
12.
Front Pharmacol ; 15: 1440147, 2024.
Article in English | MEDLINE | ID: mdl-39148534

ABSTRACT

Acrolein (ACR) is a ubiquitous environmental pollutant and byproduct of lipid peroxidation that has been implicated in male infertility. However, the molecular mechanisms underlying ACR-induced toxicity in Sertoli cells remain unclear. Given its role in inducing oxidative stress, we examined whether ferroptosis, an iron-dependent form of regulated cell death, could mediate ACR toxicity in Sertoli cells. We also tested if hydrogen sulfide (H2S), which has antioxidant and ACR detoxifying properties, could protect Sertoli cells from ACR-induced ferroptosis. ACR exposure decreased Sertoli cell viability, increased protein carbonylation and p38 MAPK phosphorylation, indicating oxidative injury. ACR also depleted glutathione (GSH), downregulated the cystine importer SLC7A11, increased intracellular ferrous iron (Fe2+) and lipid peroxidation, suggesting activation of ferroptosis. Consistently, the ferroptosis inhibitor deferoxamine (DFO) markedly attenuates ACR-induced cell death. Further studies revealed that ACR-induced ferroptotic changes were prevented by exogenous H2S and exaggerated by inhibition of endogenous H2S production. Furthermore, H2S also suppressed GPX4 inhibitor RSL3-induced intracellular ACR accumulation and ferroptosis. In summary, our study demonstrates that ACR induces ferroptotic cell death in Sertoli cells, which can be prevented by H2S through multiple mechanisms. Targeting the H2S pathway may represent a therapeutic strategy to mitigate ACR-induced Sertoli cell injury and preserve male fertility.

13.
Exp Ther Med ; 28(4): 397, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39171146

ABSTRACT

The occurrence of crystals in semen is rare, with spermine phosphate crystals being the only type commonly described. Uric acid crystal formation is significantly influenced by pH levels. The present study reported a rare case of uric acid crystals in the semen of a patient with azoospermia associated with Sertoli cell-only syndrome (SCOS). A 28-year-old male with a four-year history of primary infertility underwent clinical assessment, including a normal physical examination with small testes. Seminal fluid analysis revealed abnormal uric acid crystals. Elevated follicle-stimulating hormone, luteinizing hormone and prolactin levels were observed. The diagnosis of SCOS was confirmed through testicular sperm aspiration. Azoospermia is a medical condition characterized by the absence of sperm in the semen, specifically the absence of sperm in the pellet obtained after centrifugation. It is classified into two primary types: Obstructive and non-obstructive azoospermia. Non-obstructive azoospermia is subdivided into three categories: SCOS, hypospermatogenesis and maturation arrest. The occurrence of SCOS in azoospermic males ranges from 26.3 to 57.8%. The diagnosis of azoospermia with SCOS can be achieved through the analysis of multiple semen samples, medical history, physical examination, hormonal analysis, histopathological examination and genetic testing. The presence of uric acid crystals in seminal fluid was first reported in patients with chronic prostatitis symptoms in 2005. Despite the rarity of crystals in semen, uric acid crystals were found in the semen of an azoospermic male with SCOS.

14.
Theriogenology ; 227: 1-8, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38981313

ABSTRACT

The aim of this study was to investigate the signaling pathways involved in the proliferation and differentiation of pig Sertoli cells (SCs) mediated by thyroid hormone (T3) to provide a theoretical and practical basis for enhancing pig semen production. The effects of different concentrations of T3 on the proliferation of pig SCs were evaluated using the CCK8 assay. The impact of T3 on the proliferation and differentiation of pig SCs was further examined using RNA-seq, qPCR, and Western Blotting techniques. Additionally, the involvement of the p38 MAPK and NFκB pathways in mediating the effects of T3 on SCs proliferation and differentiation was investigated. Our findings revealed a strong correlation between the dosage of T3 and the inhibition of pig SCs proliferation and promotion of maturation. T3 regulated the activation state of the NFκB signaling pathway by upregulating IKKα, downregulating IKKß, and promoting IκB phosphorylation. Furthermore, T3 facilitated SCs maturation by upregulating AR and FSHR expression while downregulating KRT-18. In conclusion, T3 inhibits pig SCs proliferation and promote pig SCs maturation through the IKK/NFκB and p38 MAPK pathways. These findings provide valuable insights into the mechanisms by which T3 influences the proliferation and maturation of pig SCs.


Subject(s)
Cell Proliferation , NF-kappa B , Sertoli Cells , Signal Transduction , Thyroxine , p38 Mitogen-Activated Protein Kinases , Animals , Male , Swine , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Cell Proliferation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , NF-kappa B/metabolism , Signal Transduction/drug effects , Thyroxine/pharmacology , Cell Line , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , MAP Kinase Signaling System/drug effects , Cell Differentiation/drug effects
15.
Pediatr Blood Cancer ; 71(10): e31180, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39072970

ABSTRACT

Peutz-Jeghers syndrome (PJS) is a childhood-onset cancer predisposition syndrome that is associated with oral freckling and gastrointestinal polyposis. Male patients with PJS are at risk for large-cell calcifying Sertoli cell tumors in childhood. These tumors are estrogen-producing and can cause symptoms of precocious puberty, gynecomastia, and growth acceleration. Here we discuss our experience with spontaneous resolution or stabilization of breast enlargement without medical intervention in three patients with PJS and gynecomastia. These cases indicate that a watchful waiting approach can be considered in the management of gynecomastia in male children with PJS.


Subject(s)
Gynecomastia , Peutz-Jeghers Syndrome , Adolescent , Child , Child, Preschool , Humans , Male , Conservative Treatment , Gynecomastia/therapy , Gynecomastia/etiology , Peutz-Jeghers Syndrome/complications , Peutz-Jeghers Syndrome/therapy , Peutz-Jeghers Syndrome/pathology , Peutz-Jeghers Syndrome/genetics
16.
Environ Pollut ; 360: 124536, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39029862

ABSTRACT

Tri (2-Ethylhexyl) phosphate (TEHP), widely used as a fire retardant and plasticizer, has been commonly found in the environment. Its potential health-related risks, especially reproductive toxicity, have aroused concern. However, the potential cellular mechanisms remain unexplored. In this study, we aimed to investigate the molecular mechanisms underlying TEHP-caused cell damage in Sertoli cells, which play a crucial role in supporting spermatogenesis. Our findings indicate that TEHP induces apoptosis in 15P-1 mouse Sertoli cells. Subsequently, we conducted RNA sequencing analyses, which suggested that ER stress, autophagy, and MAPK-related pathways may participate in TEHP-induced cytotoxicity. Furthermore, we demonstrated that TEHP triggers ER stress, activates p38 MAPK, and inhibits autophagy flux. Then, we showed that the inhibition of ER stress or p38 MAPK activation attenuates TEHP-induced apoptosis, while the inhibition of autophagy flux is responsible for TEHP-induced apoptosis. These results collectively reveal that TEHP induces ER stress, activates p38, and inhibits autophagy flux, ultimately leading to apoptosis in Sertoli cells. These shed light on the molecular mechanisms underlying TEHP-associated testicular toxicity.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Sertoli Cells , Endoplasmic Reticulum Stress/drug effects , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Animals , Male , Autophagy/drug effects , Mice , Apoptosis/drug effects , Sequence Analysis, RNA , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Flame Retardants/toxicity , Plasticizers/toxicity , Diethylhexyl Phthalate/toxicity , Diethylhexyl Phthalate/analogs & derivatives
17.
Int J Reprod Biomed ; 22(4): 295-304, 2024 Apr.
Article in English | MEDLINE | ID: mdl-39035631

ABSTRACT

Background: Methotrexate (MET) is one of the most important chemotherapy agents used against various tumors and cancer diseases. One of the critical side effects of MET is inducing male infertility. Objective: The current study aimed to investigate Sertoli cell culture-conditioned medium (SCM) recovery effects on MET-induced conditions in rats. Materials and Methods: 30 mature male Wistar rats were randomly divided into 3 groups (n = 10). In the first group, rats received normal saline intraperitoneally. In the second group, animals received MET (10 mg/kg; intraperitoneally) once a week for 2 wk. The rats in the third group (MET+SCM) received MET and a single injection of SCM for 56 days post-MET administration. 56 days later, serum, epididymis, and testicular tissue samples were collected, and the animals were euthanized. Sperm parameters, serum levels of luteinizing hormone, follicle-stimulating hormone, and testosterone were examined. The testicular tissues were stained using hematoxylin and eosin solution, and histopathological changes were analyzed. Results: The MET-induced condition resulted in significant pathological changes in the testis, decreased hormone levels, and downregulated sperm parameters. However, SCM injection improved hormonal levels, testicular changes, and sperm parameters. Conclusion: It can be concluded that a single intra-testicular SCM injection accelerates male reproductive system recovery post-MET treatment.

18.
Biol Reprod ; 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39077996

ABSTRACT

Sertoli cells, omnipresent, somatic cells within the seminiferous tubules of the mammalian testis are essential to male fertility. Sertoli cells maintain the integrity of the testicular microenvironment, regulate hormone synthesis, and of particular importance, synthesize the active derivative of vitamin A, all trans retinoic acid (atRA), which is required for germ cell differentiation and the commitment of male germ cells to meiosis. Stages VIII-IX, when atRA synthesis occurs in the testis, coincides with multiple germ cell development and testicular restructuring events that rely on Sertoli cell gene products to proceed normally. In this study, we have synchronized and captured the mouse testis at four recurrent points of atRA synthesis to observe transcriptomic changes within Sertoli cells as mice age and the Sertoli cells are exposed to increasingly developed germ cell subtypes. This work provides comprehensive, high-resolution characterization of the timing of induction of functional Sertoli cell genes across the first wave of spermatogenesis, and outlines in silico predictions of germ cell derived signaling mechanisms targeting Sertoli cells. We have found that Sertoli cells adapt to their environment, especially to the needs of the germ cell populations present and establish germ-Sertoli cell and Sertoli-Sertoli cell junctions early, but gain many of their known immune-regulatory and protein secretory functions in preparation for spermiogenesis and spermiation. Additionally, we have found unique patterns of germ-Sertoli signaling present at each endogenous pulse of atRA, suggesting individual functions of the various germ cells in germ-Sertoli communication.

19.
Ecotoxicol Environ Saf ; 281: 116636, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917588

ABSTRACT

This study explored the regulatory role of bta-miR-149-3p in the inflammatory response induced by microcystin-leucine arginine (MC-LR) exposure in bovine Sertoli cells. The research endeavored to enhance the comprehension of the epigenetic mechanisms underlying MC-LR-induced cytotoxicity in Sertoli cells and establish a foundation for mitigating these effects in vitro. In this study, we elucidated the regulatory mechanism of bta-miR-149-3p in the MC-LR-induced inflammatory response by verifying the target gene of bta-miR-149-3p through luciferase assays and treating the cells with a bta-miR-149-3p inhibitor for 24 h. The results demonstrate that nuclear factor κB (NF-κB) acts as a downstream target gene of bta-miR-149-3p, which inhibits the MC-LR-induced inflammatory response in bovine Sertoli cells. This inhibition occurs by regulating the downregulation of tight junction constitutive proteins of the blood-testis barrier (BTB) through the suppression of the TLR-4/NF-κB signaling pathway (p < 0.05) and the up-regulation of the adhesion junction protein ß-catenin (p < 0.05). Notably, MC-LR exposure resulted in the up-regulation (p < 0.05) of inflammatory cytokines (IL-6, IL-1ß, and NLRP3) and the down-regulation (p < 0.05) of BTB tight junction constitutive proteins (ZO-1, Occludin) in Sertoli cells. Furthermore, the BTB constitutive protein ZO-1 exhibited significant down-regulation in Sertoli cells pretreated with the bta-miR-149-3p inhibitor compared to controls (p < 0.05), while Occludin showed no significant difference from CTNNB1 (p > 0.05). In summary, our findings suggest that bta-miR-149-3p suppresses the MC-LR-induced inflammatory response and alterations in the expression of BTB proteins in bovine Sertoli cells by inhibiting the TLR-4/NF-κB signaling pathway.


Subject(s)
Marine Toxins , MicroRNAs , Microcystins , NF-kappa B , Sertoli Cells , Signal Transduction , Toll-Like Receptor 4 , Animals , Cattle , Male , Microcystins/toxicity , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , NF-kappa B/metabolism , Signal Transduction/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Inflammation/chemically induced , Leucine/pharmacology
20.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892415

ABSTRACT

Elasmobranchs have an ancestral reproductive system, which offers insights into vertebrate reproductive evolution. Despite their unchanged design over 400 million years, they evolved complex mechanisms ensuring reproductive success. However, human activities induced a significant decline in elasmobranch populations worldwide. In the Mediterranean basin, the smooth-hound shark (Mustelus mustelus) is one of the species that are considered vulnerable to human activities. Conservation efforts necessitate a thorough understanding of its reproductive strategy. This study focused on mature male specimens of smooth-hound sharks that were captured in the Adriatic area and successively analyzed to provide, for the first time, a histologically detailed description of testicular development in the species. Seven phases of the spermatogenesis process were identified, along with the macromolecular characterization of cells obtained using Fourier-transform infrared imaging. Histological analysis showed structural and cellular features similar to those documented in the spermatocysts of other elasmobranchs. The examination of the evolution and migration of both germinative and Sertoli cells at each phase revealed their close connection. Furthermore, different expression levels of lipids, proteins, and phosphates (DNA) at each spermatogenesis stage were observed. This research provided new information on spermatogenesis in the common smooth-hound shark, which is crucial for conservation efforts against population decline and anthropogenic pressures.


Subject(s)
Sharks , Spermatogenesis , Testis , Animals , Sharks/metabolism , Male , Testis/cytology , Testis/metabolism , Sertoli Cells/metabolism , Sertoli Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL