Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
1.
Part Fibre Toxicol ; 21(1): 30, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39118174

ABSTRACT

Plastic pollution is an emerging environmental issue, with microplastics and nanoplastics raising health concerns due to bioaccumulation. This work explored the impact of polystyrene nanoparticle (PS-NPs) exposure during prepuberty on male reproductive function post maturation in rats. Rats were gavaged with PS-NPs (80 nm) at 0, 3, 6, 12 mg/kg/day from postnatal day 21 to 95. PS-NPs accumulated in the testes and reduced sperm quality, serum reproductive hormones, and testicular coefficients. HE staining showed impaired spermatogenesis. PS-NPs disrupted the blood-testis barrier (BTB) by decreasing junction proteins, inducing inflammation and apoptosis. Transcriptomics identified differentially expressed genes related to metabolism, lysosome, apoptosis, and TLR4 signaling. Molecular docking revealed Cordycepin could compete with polystyrene for binding to TLR4. Cordycepin alleviated oxidative stress and improved barrier function in PS-NPs treated Sertoli cells. In conclusion, prepubertal PS-NPs exposure induces long-term reproductive toxicity in male rats, likely by disrupting spermatogenesis through oxidative stress and BTB damage. Cordycepin could potentially antagonize this effect by targeting TLR4 and warrants further study as a protective agent. This study elucidates the mechanisms underlying reproductive toxicity of PS-NPs and explores therapeutic strategies.


Subject(s)
Blood-Testis Barrier , Deoxyadenosines , Nanoparticles , Polystyrenes , Spermatogenesis , Testis , Animals , Male , Deoxyadenosines/pharmacology , Blood-Testis Barrier/drug effects , Polystyrenes/toxicity , Nanoparticles/toxicity , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Molecular Docking Simulation , Microplastics/toxicity , Toll-Like Receptor 4/metabolism , Apoptosis/drug effects , Sexual Maturation/drug effects , Protective Agents/pharmacology
2.
Pestic Biochem Physiol ; 203: 106010, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39084803

ABSTRACT

Thiram, a prevalent dithiocarbamate insecticide in agriculture, is widely employed as a crop insecticide and preservative. Chronic exposure to thiram has been linked to various irreversible damages, including tibial cartilage dysplasia, erythrocytotoxicity, renal issues, and immune system compromise. Limited research exists on its effects on reproductive organs. This study investigated the reproductive toxicology in mouse testes exposure to varying concentrations (0, 30, 60, and 120 mg/kg) of thiram. Our study uncovered a series of adverse effects in mice subjected to thiram exposure, including emaciation, stunted growth, decreased water intake, and postponed testicular maturation. Biochemical analysis in thiram-exposed mice showed elevated levels of LDH and AST, while ALP, TG, ALT, and urea were decreased. Histologically, thiram disrupted the testis' microarchitecture and compromised its barrier function by widening the gap between spermatogenic cells and promoting fibrosis. The expression of pro-apoptotic genes (Bax, APAF1, Cytc, and Caspase-3) was downregulated, whereas Bcl-2 expression increased in thiram-treated mice compared to controls. Conversely, the expression of Atg5 was upregulated, and mTOR and p62 expression decreased, with a trend towards lower LC3b levels. Thiram also disrupted the blood-testis barrier, significantly reducing the mRNA expression of zona occludens-1 (ZO-1) and occludin. In conclusion, chronic exposure to high thiram concentrations (120 mg/kg) caused testicular tissue damage, affecting the blood-testis barrier and modulating apoptosis and autophagy through the Bcl-2/Bax and mTOR/Atg5/p62 pathways. This study contributes to understanding the molecular basis of thiram-induced reproductive toxicity and underscores the need for further research and precautions for those chronically exposed to thiram and its environmental residuals.


Subject(s)
Apoptosis , Autophagy-Related Protein 5 , Autophagy , Blood-Testis Barrier , Proto-Oncogene Proteins c-bcl-2 , TOR Serine-Threonine Kinases , Testis , Thiram , bcl-2-Associated X Protein , Animals , Male , Apoptosis/drug effects , Mice , TOR Serine-Threonine Kinases/metabolism , Blood-Testis Barrier/drug effects , Testis/drug effects , Testis/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics , Autophagy/drug effects , Thiram/toxicity , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Insecticides/toxicity , Signal Transduction/drug effects
3.
Toxicology ; 507: 153888, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39019315

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) can reduce sperm number, but the mechanisms of defective spermatogenesis induced by TiO2 NPs have not been studied through cell-cell interactions at present. A kind of biomimetic three-dimensional blood-testis barrier microfluidic chip capable of intercellular communication was constructed with soft lithography techniques, including Sertoli cell (TM4), spermatogonia (GC-1) and vascular endothelial cell units, to study the mechanisms of TiO2 NPs-induced defective spermatogenesis. TM4 and GC-1 cells cultured in TiO2 NPs exposure and control chips were collected for transcriptomics and metabonomics analysis, and key proteins and metabolites in changed biological processes were validated. In TM4 cells, TiO2 NPs suppressed glucose metabolism, especially lactate production, which reduced energy substrate supply for spermatogenesis. TiO2 NPs also decreased the levels of key proteins and metabolites of lactate production. In GC-1 cells, TiO2 NPs disturbed chemokine signaling pathways regulating cell proliferation and interfered with glutathione metabolism. The Cxcl13, Stat3 and p-Stat3 levels and cell proliferation rate were decreased, and the GSR, GPX4 and GSH contents were increased in GC-1 cells in chips under TiO2 NPs treatment. The decrease in energy substrate supply for spermatogenesis and inhibition of spermatogonia proliferation could be the main mechanisms of defective spermatogenesis induced by TiO2 NPs.


Subject(s)
Blood-Testis Barrier , Sertoli Cells , Spermatogenesis , Spermatogonia , Titanium , Male , Titanium/toxicity , Spermatogenesis/drug effects , Animals , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Blood-Testis Barrier/drug effects , Mice , Spermatogonia/drug effects , Spermatogonia/metabolism , Spermatogonia/pathology , Cell Line , Metal Nanoparticles/toxicity , Lab-On-A-Chip Devices , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Cell Communication/drug effects
4.
Sci Total Environ ; 948: 174738, 2024 Oct 20.
Article in English | MEDLINE | ID: mdl-39009145

ABSTRACT

2,2',4,4'-Tetrabromodiphenyl ether (PBDE-47), being the most prevalent congener of polybrominated diphenyl ethers (PBDEs), has been found to accumulate greatly in the environment and induce spermatogenesis dysfunction. However, the specific underlying factors and mechanisms have not been elucidated. Herein, male Sprague-Dawley (SD) rats were exposed to corn oil, 10 mg/kg body weight (bw) PBDE-47 or 20 mg/kg bw PBDE-47 by gavage for 30 days. PBDE-47 exposure led to blood-testis barrier (BTB) integrity disruption and aberrant spermatogenesis. Given that Sertoli cells are the main toxicant target, to explore the potential mechanism involved, we performed RNA sequencing (RNA-seq) in Sertoli cells, and the differentially expressed genes were shown to be enriched in ferroptosis and lysosomal pathways. We subsequently demonstrated that ferroptosis was obviously increased in testes and Sertoli cells upon exposure to PBDE-47, and the junctional function of Sertoli cells was restored after treatment with the ferroptosis inhibitor ferrostatin-1. Since glutathione peroxidase 4 (GPX4) was dramatically reduced in PBDE-47-exposed testes and Sertoli cells and considering the RNA-sequencing results, we examined the activity of chaperone-mediated autophagy (CMA) and verified that the expression of LAMP2a and HSC70 was upregulated significantly after PBDE-47 exposure. Notably, Lamp2a knockdown not only inhibited ferroptosis by suppressing GPX4 degradation but also restored the impaired junctional function induced by PBDE-47. These collective findings strongly indicate that PBDE-47 induces Sertoli cell ferroptosis through CMA-mediated GPX4 degradation, resulting in decreased BTB-associated protein expression and eventually leading to BTB integrity disruption and spermatogenesis dysfunction.


Subject(s)
Blood-Testis Barrier , Ferroptosis , Halogenated Diphenyl Ethers , Animals , Male , Rats , Blood-Testis Barrier/drug effects , Ferroptosis/drug effects , Halogenated Diphenyl Ethers/toxicity , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Testis/drug effects
5.
Lipids Health Dis ; 23(1): 180, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862993

ABSTRACT

BACKGROUND: The management of male infertility continues to encounter an array of challenges and constraints, necessitating an in-depth exploration of novel therapeutic targets to enhance its efficacy. As an eight-carbon medium-chain fatty acid, octanoic acid (OCA) shows promise for improving health, yet its impact on spermatogenesis remains inadequately researched. METHODS: Mass spectrometry was performed to determine the fatty acid content and screen for a pivotal lipid component in the serum of patients with severe spermatogenesis disorders. The sperm quality was examined, and histopathological analysis and biotin tracer tests were performed to assess spermatogenesis function and the integrity of the blood-testis barrier (BTB) in vivo. Cell-based in vitro experiments were carried out to investigate the effects of OCA administration on Sertoli cell dysfunction. This research aimed to elucidate the mechanism by which OCA may influence the function of Sertoli cells. RESULTS: A pronounced reduction in OCA content was observed in the serum of patients with severe spermatogenesis disorders, indicating that OCA deficiency is related to spermatogenic disorders. The protective effect of OCA on reproduction was tested in a mouse model of spermatogenic disorder induced by busulfan at a dose 30 mg/kg body weight (BW). The mice in the study were separated into distinct groups and administered varying amounts of OCA, specifically at doses of 32, 64, 128, and 256 mg/kg BW. After evaluating sperm parameters, the most effective dose was determined to be 32 mg/kg BW. In vivo experiments showed that treatment with OCA significantly improved sperm quality, testicular histopathology and BTB integrity, which were damaged by busulfan. Moreover, OCA intervention reduced busulfan-induced oxidative stress and autophagy in mouse testes. In vitro, OCA pretreatment (100 µM) significantly ameliorated Sertoli cell dysfunction by alleviating busulfan (800 µM)-induced oxidative stress and autophagy. Moreover, rapamycin (5 µM)-induced autophagy led to Sertoli cell barrier dysfunction, while OCA administration exerted a protective effect by alleviating autophagy. CONCLUSIONS: This study demonstrated that OCA administration suppressed oxidative stress and autophagy to alleviate busulfan-induced BTB damage. These findings provide a deeper understanding of the toxicology of busulfan and a promising avenue for the development of novel OCA-based therapies for male infertility.


Subject(s)
Autophagy , Blood-Testis Barrier , Busulfan , Caprylates , Oxidative Stress , Sertoli Cells , Spermatogenesis , Male , Animals , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Busulfan/adverse effects , Caprylates/pharmacology , Oxidative Stress/drug effects , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Humans , Spermatogenesis/drug effects , Autophagy/drug effects , Infertility, Male/drug therapy , Infertility, Male/chemically induced , Infertility, Male/pathology , Testis/drug effects , Testis/pathology , Testis/metabolism , Spermatozoa/drug effects , Spermatozoa/metabolism , Adult
6.
Environ Geochem Health ; 46(7): 238, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849627

ABSTRACT

Microplastics (MPs) are defined as plastic particles or fragments with a diameter of less than 5 mm. These particles have been identified as causing male reproductive toxicity, although the precise mechanism behind this association is yet to be fully understood. Recent research has found that exposure to polystyrene microplastics (PS-MPs) can disrupt spermatogenesis by impacting the integrity of the blood-testis barrier (BTB), a formidable barrier within mammalian blood tissues. The BTB safeguards germ cells from harmful substances and infiltration by immune cells. However, the disruption of the BTB leads to the entry of environmental pollutants and immune cells into the seminiferous tubules, resulting in adverse reproductive effects. Additionally, PS-MPs induce reproductive damage by generating oxidative stress, inflammation, autophagy, and alterations in the composition of intestinal flora. Despite these findings, the precise mechanism by which PS-MPs disrupt the BTB remains inconclusive, necessitating further investigation into the underlying processes. This review aims to enhance our understanding of the pernicious effects of PS-MP exposure on the BTB and explore potential mechanisms to offer novel perspectives on BTB damage caused by PS-MPs.


Subject(s)
Blood-Testis Barrier , Microplastics , Polystyrenes , Microplastics/toxicity , Polystyrenes/toxicity , Male , Humans , Blood-Testis Barrier/drug effects , Animals , Spermatogenesis/drug effects , Oxidative Stress/drug effects , Environmental Pollutants/toxicity
7.
Ecotoxicol Environ Saf ; 280: 116578, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38861803

ABSTRACT

Sertoli cells (SCs) maintain testicular homeostasis and promote spermatogenesis by forming the blood-testis barrier (BTB) and secreting growth factors. The pro-proliferative and anti-apoptotic effects of nerve growth factor (NGF) on SCs have been proved previously. It is still unclear whether the damage effect of arsenic on testis is related to the inhibition of NGF expression, and whether NGF can mitigate arsenic-induced testicular damage by decreasing the damage of SCs induced by arsenic. Here, the lower expression of NGF in testes of arsenic exposed mice (freely drinking water containing 15 mg/l of NaAsO2) was observed through detection of Western blot and Real-time PCR. Subsequently, hematoxylin and eosin (HE) staining, Evans blue staining and transmission electron microscopy were used to evaluate the pathology, BTB permeability and tight junction integrity in testes of control mice, arsenic exposed mice (freely drinking water containing 15 mg/l of NaAsO2) and arsenic + NGF treated mice (freely drinking water containing 15 mg/l of NaAsO2 + intraperitoneal injection with 30 µg/kg of NGF), respectively. Evidently, spermatogenic tubule epithelial cells in testis of arsenic exposed mice were disordered and the number of cell layers was reduced, accompanied by increased permeability and damaged integrity of the tight junction in BTB, but these changes were less obvious in testes of mice treated with arsenic + NGF. In addition, the sperm count, motility and malformation rate of mice treated with arsenic + NGF were also improved. On the basis of the above experiments, the viability and apoptosis of primary cultured SCs treated with arsenic (10 µM NaAsO2) or arsenic + NGF (10 µM NaAsO2 + 100 ng/mL NGF) were detected by Cell counting kit-8 (CCK8) and transferase-mediated DUTP-biotin nick end labeling (TUNEL) staining, respectively. It is found that NGF ameliorated the decline of growth activity and the increase of apoptosis in arsenic-induced SCs. This remarkable biological effect that NGF inhibited the increase of Bax expression and the decrease of Bcl-2 expression in arsenic-induced SCs was also determined by western blot and Real-time PCR. Moreover, the decrease in transmembrane resistance (TEER) and the expression of tight junction proteins ZO-1 and occludin was mitigated in SCs induced by arsenic due to NGF treatment. In conclusion, the above results confirmed that NGF could ameliorate the injury effects of arsenic on testis, which might be related to the function of NGF to inhibit arsenic-induced SCs injury.


Subject(s)
Arsenic , Blood-Testis Barrier , Nerve Growth Factor , Sertoli Cells , Testis , Animals , Male , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Mice , Arsenic/toxicity , Testis/drug effects , Testis/pathology , Blood-Testis Barrier/drug effects , Spermatogenesis/drug effects , Apoptosis/drug effects , Tight Junctions/drug effects
8.
Am J Physiol Cell Physiol ; 327(2): C291-C309, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38826136

ABSTRACT

Per- and polyfluoroalkyl substances (PFASs) are a family of "forever chemicals" including perfluorooctane sulfonate (PFOS). These toxic chemicals do not break down in the environment or in our bodies. In the human body, PFOS and perfluoroctanoic acid (PFOA) have a half-life (T1/2) of about 4-5 yr so low daily consumption of these chemicals can accumulate in the human body to a harmful level over a long period. Although the use of PFOS in consumer products was banned in the United States in 2022/2023, this forever chemical remains detectable in our tap water and food products. Every American tested has a high level of PFAS in their blood (https://cleanwater.org/pfas-forever-chemicals). In this report, we used a Sertoli cell blood-testis barrier (BTB) model with primary Sertoli cells cultured in vitro with an established functional tight junction (TJ)-permeability barrier that mimicked the BTB in vivo. Treatment of Sertoli cells with PFOS was found to perturb the TJ-barrier, which was the result of cytoskeletal disruption across the cell cytoplasm, disrupting actin and microtubule polymerization. These changes thus affected the proper localization of BTB-associated proteins at the BTB. Using RNA-Seq transcriptome profiling, bioinformatics analysis, and pertinent biochemical and cell biology techniques, it was discovered that PFOS -induced Sertoli cell toxicity through the c-Jun N-terminal kinase (JNK; also known as stress-activated protein kinase, SAPK) and its phosphorylated/active form p-JNK signaling pathway. More importantly, KB-R7943 mesylate (KB), a JNK/p-JNK activator, was capable of blocking PFOS-induced Sertoli cell injury, supporting the notion that PFOS-induced cell injury can possibly be therapeutically managed.NEW & NOTEWORTHY PFOS induces Sertoli cell injury, including disruption of the 1) blood-testis barrier function and 2) cytoskeletal organization, which, in turn, impedes male reproductive function. These changes are mediated by JNK/p-JNK signaling pathway. However, the use of KB-R7943, a JNK/p-JNK activator was capable of blocking PFOS-induced Sertoli cell injury, supporting the possibility of therapeutically managing PFOS-induced reproductive dysfunction.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , JNK Mitogen-Activated Protein Kinases , Sertoli Cells , Fluorocarbons/toxicity , Alkanesulfonic Acids/toxicity , Male , Animals , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , RNA-Seq , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , Cells, Cultured , Mice , Rats , Rats, Sprague-Dawley
9.
Reprod Toxicol ; 128: 108633, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38944211

ABSTRACT

3-chloro-1,2-propanediol (3-MCPD) is a newly discovered food process pollutant with nephrotoxicity. And the mechanism by which 3-MCPD affects male spermatogenesis has not been fully studied. Cell viability, blood-testis barrier (BTB) related protein, progesterone content, reactive oxygen species (ROS) generation, and cell apoptosis were determined by a CCK8 assay, western blot, ELISA, flow cytometry, and TUNEL staining, respectively. Wistar rats were divided into three groups: low-dose 3-MCPD, high-dose 3-MCPD, and control. Sperm parameters, hormonal levels, and biomarkers of oxidative stress in the testis and epididymis were detected by ELISA. Multiple molecular experiments including molecular docking and western blot were used to elucidate the underlying mechanisms. 3-MCPD affects testicular cell activity, and promotes ROS production and apoptosis. Disrupting the integrity of BTB in the body, downregulating sex hormones and sperm quality, and promoting apoptosis. 3-MCPD may function through CYP2C9. This study preliminarily explores the mechanism by which 3-MCPD affects spermatogenesis. It was found that 3-MCPD destroys the structure and function of BTB and damages the testicular function of male mice, thus affecting the process of spermatogenesis via CYP2C9.


Subject(s)
Apoptosis , Oxidative Stress , Rats, Wistar , Reactive Oxygen Species , Testis , alpha-Chlorohydrin , Animals , Male , Oxidative Stress/drug effects , alpha-Chlorohydrin/toxicity , alpha-Chlorohydrin/analogs & derivatives , Testis/drug effects , Testis/pathology , Testis/metabolism , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Spermatogenesis/drug effects , Spermatozoa/drug effects , Infertility, Male/chemically induced , Rats , Blood-Testis Barrier/drug effects , Molecular Docking Simulation
10.
Ecotoxicol Environ Saf ; 279: 116502, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38788563

ABSTRACT

BACKGROUND: Despite the known reproductive toxicity induced by triptolide (TP) exposure, the regulatory mechanism underlying testicular vacuolization injury caused by TP remains largely obscure. METHODS: Male mice were subjected to TP at doses of 15, 30, and 60 µg/kg for 35 consecutive days. Primary Sertoli cells were isolated from 20-day-old rat testes and exposed to TP at concentrations of 0, 40, 80, 160, 320, and 640 nM. A Biotin tracer assay was conducted to assess the integrity of the blood-testis barrier (BTB). Transepithelial electrical resistance (TER) assays were employed to investigate BTB function in primary Sertoli cells. Histological structures of the testes and epididymides were stained with hematoxylin and eosin (H&E). The expression and localization of relevant proteins or pathways were assessed through Western blotting or immunofluorescence staining. RESULTS: TP exposure led to dose-dependent testicular injuries, characterized by a decreased organ coefficient, reduced sperm concentration, and the formation of vacuolization damage. Furthermore, TP exposure disrupted BTB integrity by reducing the expression levels of tight junction (TJ) proteins in the testes without affecting basal ectoplasmic specialization (basal ES) proteins. Through the TER assay, we identified that a TP concentration of 160 nM was optimal for elucidating BTB function in primary Sertoli cells, correlating with reductions in TJ protein expression. Moreover, TP exposure induced changes in the distribution of the BTB and cytoskeleton-associated proteins in primary Sertoli cells. By activating the AKT/mTOR signaling pathway, TP exposure disturbed the balance between mTORC1 and mTORC2, ultimately compromising BTB integrity in Sertoli cells. CONCLUSION: This investigation sheds light on the impacts of TP exposure on testes, elucidating the mechanism by which TP exposure leads to testicular vacuolization injury and offering valuable insights into comprehending the toxic effects of TP exposure on testes.


Subject(s)
Blood-Testis Barrier , Cytoskeleton , Diterpenes , Epoxy Compounds , Phenanthrenes , Proto-Oncogene Proteins c-akt , Sertoli Cells , Signal Transduction , TOR Serine-Threonine Kinases , Testis , Male , Animals , Sertoli Cells/drug effects , Sertoli Cells/pathology , Diterpenes/toxicity , Phenanthrenes/toxicity , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Testis/drug effects , Testis/pathology , Epoxy Compounds/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Mice , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/pathology , Cytoskeleton/drug effects , Rats , Vacuoles/drug effects , Rats, Sprague-Dawley
11.
Toxicol Sci ; 200(1): 70-78, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38565259

ABSTRACT

Peritubular macrophages (PTMφ) are predominantly localized near spermatogonial stem cells in the testis. We previously revealed that exposure of peripubertal male Fischer rats to mono-(2-ethylhexyl) phthalate (MEHP) leads to increased PTMφs in the testis. The mechanisms that trigger increases in PTMφs in the testis are poorly understood. However, MEHP exposure is known to both induce spermatocyte apoptosis and to perturb the blood-testis barrier (BTB). This study aims to elucidate the association between the disruption of BTB and the increases of PTMφs in the testis by comparing the effects observed with MEHP to 2 other testicular toxicants with variable effects on the BTB and subtype of germ cell undergoing apoptosis. Methoxyacetic acid (MAA) acts directly on spermatocytes and does not affect BTB function, whereas cadmium chloride (CdCl2) induces profound injury to BTB. The results indicated that MAA exposure significantly increased spermatocyte apoptosis, whereas no significant changes in the numbers of PTMφs in the testis occurred. In contrast, CdCl2 exposure disrupted BTB function and increased the abundance of PTMφs in the testis. To further investigate whether MEHP-induced changes in BTB integrity accounted for the increase in PTMφs, a plasmid for LG3/4/5, the functional component of laminin-alpha 2, was overexpressed in the testis to stabilize BTB integrity before MEHP exposure. The results showed that LG3/4/5 overexpression substantially reduced the ability of MEHP to compromise BTB integrity and prevented the increase in PTMφ numbers after MEHP exposure. These results indicate that BTB disruption is necessary to increase PTMφs in the testis induced by toxicants.


Subject(s)
Apoptosis , Blood-Testis Barrier , Diethylhexyl Phthalate , Macrophages , Rats, Inbred F344 , Testis , Animals , Male , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/pathology , Blood-Testis Barrier/metabolism , Diethylhexyl Phthalate/toxicity , Diethylhexyl Phthalate/analogs & derivatives , Testis/drug effects , Testis/pathology , Testis/metabolism , Macrophages/drug effects , Apoptosis/drug effects , Cadmium Chloride/toxicity , Acetates/toxicity , Rats , Spermatocytes/drug effects , Spermatocytes/pathology
12.
J Hazard Mater ; 470: 134126, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38554509

ABSTRACT

Cadmium (Cd) is a well-known testis toxicant. The blood-testis barrier (BTB) is a crucial component of the testis. Cd can disrupt the integrity of the BTB and reproductive function. However, the mechanism of Cd-induced disruption of BTB and testicular damage has not been fully elucidated. Here, our study investigates the effects of Cd on BTB integrity and testicular dysfunction. 80 (aged 1 day) Hy-Line white variety chickens were randomly designed into 4 groups and treated for 90 days, as follows: control group (essential diet), 35 Cd, 70 Cd and 140 Cd groups (35, 70 and 140 mg/kg Cd). The results found that Cd exposure diminished volume of the testes and induced histopathological lesions in the testes. Exposure to Cd induced an inflammatory response, disrupted the structure and function of the FAK/occludin/ZO-1 protein complex and disrupted the tight junction and adherens junction in the BTB. In addition, Cd exposure reduced the expression of steroid-related proteins and inhibited testosterone synthesis. Taken together, these data elucidate that Cd disrupts the integrity of the BTB and further inhibits spermatogenesis by dissociating the FAK/occludin/ZO-1 complex, which provides a basis for further investigation into the mechanisms of Cd-induced impairment of male reproductive function and pharmacological protection.


Subject(s)
Blood-Testis Barrier , Cadmium , Chickens , Testis , Animals , Male , Blood-Testis Barrier/drug effects , Cadmium/toxicity , Focal Adhesion Kinase 1/metabolism , Occludin/metabolism , Spermatogenesis/drug effects , Testis/drug effects , Testis/metabolism , Testis/pathology , Testosterone/blood , Zonula Occludens-1 Protein/metabolism
13.
Acta Pharmacol Sin ; 45(6): 1237-1251, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38472317

ABSTRACT

Both epidemiological and animal studies suggest that adverse environment during pregnancy can change the offspring development programming, but it is difficult to achieve prenatal early warning. In this study we investigated the impact of prenatal dexamethasone exposure (PDE) on sperm quality and function of blood-testis barrier (BTB) in adult offspring and the underlying mechanisms. Pregnant rats were injected with dexamethasone (0.1, 0.2 and 0.4 mg·kg-1·d-1, s.c.) from GD9 to GD20. After weaning (PW4), the pups were fed with lab chow. At PW12 and PW28, the male offspring were euthanized to collect blood and testes samples. We showed that PDE significantly decreased sperm quality (including quantity and motility) in male offspring, which was associated with impaired BTB and decreased CX43/E-cadherin expression in the testis. We demonstrated that PDE induced morphological abnormalities of fetal testicle and Sertoli cell development originated from intrauterine. By tracing to fetal testicular Sertoli cells, we found that PDE dose-dependently increased expression of histone lysine demethylases (KDM1B), decreasing histone 3 lysine 9 dimethylation (H3K9me2) levels of follistatin-like-3 (FSTL3) promoter region and increased FSTL3 expression, and inhibited TGFß signaling and CX43/E-cadherin expression in offspring before and after birth. These results were validated in TM4 Sertoli cells following dexamethasone treatment. Meanwhile, the H3K9me2 levels of FSTL3 promoter in maternal peripheral blood mononuclear cell (PBMC) and placenta were decreased and its expression increased, which was positively correlated with the changes in offspring testis. Based on analysis of human samples, we found that the H3K9me2 levels of FSTL3 promoter in maternal blood PBMC and placenta were positively correlated with fetal blood testosterone levels after prenatal dexamethasone exposure. We conclude that PDE can reduce sperm quality in adult offspring rats, which is related to the damage of testis BTB via epigenetic modification and change of FSTL3 expression in Sertoli cells. The H3K9me2 levels of the FSTL3 promoter and its expression in the maternal blood PBMC can be used as a prenatal warning marker for fetal testicular dysplasia.


Subject(s)
Blood-Testis Barrier , Dexamethasone , Prenatal Exposure Delayed Effects , Signal Transduction , Animals , Male , Female , Pregnancy , Dexamethasone/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Signal Transduction/drug effects , Rats , Spermatozoa/drug effects , Spermatozoa/metabolism , Transforming Growth Factor beta/metabolism , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testis/drug effects , Testis/metabolism , Testis/pathology
14.
Asian J Androl ; 26(3): 295-301, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38284772

ABSTRACT

Cuproptosis, a novel mechanism of programmed cell death, has not been fully explored in the context of spermatogenic cells. This study investigated the potential involvement of cuproptosis in spermatogenic cell death using a mouse model of copper overload. Sixty male Institute of Cancer Research (ICR) mice were randomly divided into four groups that received daily oral gavage with sodium chloride (control) or copper sulfate (CuSO 4 ) at 50 mg kg -1 , 100 mg kg -1 , or 200 mg kg -1 , for 42 consecutive days. Mice subjected to copper overload exhibited a disruption in copper homeostasis. Additionally, significant upregulated expression of key cuproptosis factors was accompanied by a significant rise in the rates of testicular tissue cell apoptosis. Immunohistochemical analysis revealed the presence of ferredoxin 1 (Fdx1) in Sertoli cells, Leydig cells, and spermatogenic cells at various stages of testicular development, and the Fdx1-positive staining area was significantly increased in copper-overloaded mice. Mitochondrial dysfunction and decreased adenosine triphosphate levels were also observed, further implicating mitochondrial damage under cuproptosis. Further analyses revealed pathological lesions and blood-testis barrier destruction in the testicular tissue, accompanied by decreased sperm concentration and motility, in copper-overloaded mice. In summary, our results indicate that copper-overloaded mice exhibit copper homeostasis disorder in the testicular tissue and that cuproptosis participates in spermatogenic cell death. These findings provide novel insights into the pathogenic mechanisms underlying spermatogenic cell death and provide initial experimental evidence for the occurrence of cuproptosis in the testis.


Subject(s)
Apoptosis , Copper , Sertoli Cells , Spermatogenesis , Testis , Animals , Male , Mice , Testis/pathology , Testis/drug effects , Testis/metabolism , Apoptosis/drug effects , Copper/toxicity , Sertoli Cells/drug effects , Sertoli Cells/pathology , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Mice, Inbred ICR , Ferredoxins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Copper Sulfate/toxicity , Copper Sulfate/pharmacology , Leydig Cells/drug effects , Leydig Cells/pathology , Leydig Cells/metabolism , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/pathology , Blood-Testis Barrier/metabolism , Cell Death/drug effects , Adenosine Triphosphate/metabolism
15.
Andrology ; 10(2): 377-391, 2022 02.
Article in English | MEDLINE | ID: mdl-34535976

ABSTRACT

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


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Blood-Testis Barrier/drug effects , Busulfan/adverse effects , Infertility, Male/chemically induced , Spermatogenesis/drug effects , Animals , Autoantigens/metabolism , Cell Membrane Permeability/drug effects , Collagen Type IV/metabolism , Disease Models, Animal , Male , Matrix Metalloproteinase 9/metabolism , Rats , Seminiferous Epithelium/metabolism
16.
Toxins (Basel) ; 13(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34941713

ABSTRACT

Zearalenone (ZEA), a common mycotoxin in grains and animal feeds, has been associated with male reproductive disorders. However, the potential toxicity mechanism of ZEA is not fully understood. In this study, in vivo and in vitro models were used to explore the effects of ZEA on the blood-testis barrier (BTB) and related molecular mechanisms. First, male BALB/C mice were administered ZEA orally (40 mg/kg·bw) for 5-7 d. Sperm motility, testicular morphology, and expressions of BTB junction proteins and autophagy-related proteins were evaluated. In addition, TM4 cells (mouse Sertoli cells line) were used to delineate the molecular mechanisms that mediate the effects of ZEA on BTB. Our results demonstrated that ZEA exposure induced severe testicular damage in histomorphology and an ultrastructural, time-dependent decrease in the expression of blood-testis barrier junction-related proteins, accompanied by an increase in the expression of autophagy-related proteins. Additionally, similar to the in vitro results, the dose-dependent treatment of ZEA increased the level of cytoplasmic Ca2+ and the levels of the autophagy markers LC3-II and p62, in conjunction with a decrease in the BTB junction proteins occludin, claudin-11, and Cx43, with the dislocation of the gap junction protein Cx43. Meanwhile, inhibition of autophagy by CQ and 3-MA or inhibition of cytoplasmic Ca2+ by BAPTA-AM was sufficient to reduce the effects of ZEA on the TM4 cell BTB. To summarize, this study emphasizes the role of Ca2+-mediated autophagy in ZEA-induced BTB destruction, which deepens our understanding of the molecular mechanism of ZEA-induced male reproductive disorders.


Subject(s)
Blood-Testis Barrier/drug effects , Mycotoxins/toxicity , Myelin and Lymphocyte-Associated Proteolipid Proteins/drug effects , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Sperm Motility/drug effects , Zearalenone/metabolism , Zearalenone/toxicity , Animals , Autophagy/drug effects , Male , Mice , Mice, Inbred BALB C , Mycotoxins/metabolism , Sertoli Cells/drug effects , Testis/drug effects
17.
Aging (Albany NY) ; 13(23): 25440-25452, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34904960

ABSTRACT

As one of the most commonly used nanoparticles, titanium dioxide nanoparticles (TiO2-NPs) are widely used as coating reagents in cosmetics, medicine and other industries. The increasing risk of exposure to TiO2-NPs raises concerns about their safety. In this study, we investigated the mechanism by which TiO2-NPs cross the blood-testis barrier (BTB). TM-4 cells were selected as an in vitro Sertoli cell model of BTB. Cell viability, cell morphological changes, apoptosis, oxidative damage, and the expression levels of actin regulatory and tight junction (TJ) proteins were assessed in TM-4 cells treated with 3-nm and 24-nm TiO2-NPs. Cells treated with 3-nm TiO2-NPs exhibited increased cytotoxicity and decreased Annexin II expression, whereas cells treated with 24-nm TiO2-NPs exhibited increased Arp 3 and c-Src expression. Both TiO2-NPs induced significant oxidative stress, decreased the expression of TJ proteins (occludin, ZO-1 and claudin 5), damaged the TJ structure, and exhibited enlarged gaps between TM-4 cells. Our results indicated that both TiO2-NPs crossed the BTB by disrupting actin-based adhesive junctions of TM-4 cells; however, apoptosis was not observed. Our results provide new insights into how TiO2-NPs cross the BTB.


Subject(s)
Actins/antagonists & inhibitors , Blood-Testis Barrier/drug effects , Cell Adhesion/drug effects , Metal Nanoparticles/adverse effects , Titanium/adverse effects , Actins/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Male , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Tight Junction Proteins/metabolism
18.
Cells ; 10(9)2021 09 13.
Article in English | MEDLINE | ID: mdl-34572053

ABSTRACT

A protocol for the isolation and long-term propagation of adult rat Sertoli cells (SCs) using conditional reprogramming (CR) was developed and the formation of tight junctions as an in vitro model for the blood testis barrier (BTB) was studied. Three pure primary SC lines were isolated successfully and maintained for several months without significant changes in expression levels of SC-typical markers such as SRY-box transcription factor 9 (SOX9), transferrin, clusterin, androgen receptor (AR), and GATA binding protein 1 (GATA1). In addition to AR expression, the tight junction proteins, zonula occludens-1 (ZO-1) and the junctional adhesion molecule-3 (JAM-3), were upregulated and the SC barrier integrity was enhanced by testosterone. Peritubular/myoid cells did not increase the tightness of the SC. The cytokines, interleukin-6 (IL-6), bone morphogenetic protein-2 (BMP2), and transforming growth factor beta-3 (TGF-ß3), negatively affected the tightness of the SC barrier. We have established a protocol for the isolation and long-term propagation of highly pure primary adult rat SCs, which are able to respond to androgen treatments, to form tight junctions and to maintain the mRNA expression of SC-specific genes. By applying this new method, adult SCs can now be analyzed in more detail and might serve as an in vitro model for the study of many SC functions.


Subject(s)
Androgens/pharmacology , Biomarkers/metabolism , Blood-Testis Barrier/physiology , Gene Expression Regulation , Sertoli Cells/cytology , Testis/cytology , Animals , Blood-Testis Barrier/drug effects , Cytokines/metabolism , Male , Rats , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testis/drug effects , Testis/metabolism , Tight Junctions/drug effects , Tight Junctions/physiology
19.
Reprod Biol ; 21(4): 100562, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34555686

ABSTRACT

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


Subject(s)
Estrogens/deficiency , Intercellular Junctions/metabolism , Isoflavones/pharmacology , Membrane Proteins/metabolism , Seminiferous Epithelium/metabolism , Animals , Blood-Testis Barrier/drug effects , Female , Glycation End Products, Advanced/metabolism , Letrozole , Male , Maternal Exposure , Pregnancy , Prenatal Exposure Delayed Effects , Rats, Wistar , Seminiferous Epithelium/drug effects , Sexual Maturation/drug effects
20.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34453520

ABSTRACT

The human testis can be infected by a large number of RNA and DNA viruses. While various RNA virus infections may induce orchitis and impair testicular functions, DNA virus infection rarely affects the testis. Mechanisms underlying the differential effects of RNA and DNA viral infections on the testis remain unclear. In the current study, we therefore examined the effects of viral RNA and DNA sensor signaling pathways on mouse Sertoli cells (SC) and Leydig cells (LC). The local injection of viral RNA analogue polyinosinic-polycytidylic acid [poly(I:C)] into the testis markedly disrupted spermatogenesis, whereas the injection of the herpes simplex virus (HSV) DNA analogue HSV60 did not affect spermatogenesis. Poly(I:C) dramatically induced the expression of the proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 in SC and LC through Toll-like receptor 3 and interferon ß promoter stimulator 1 signaling pathways, impairing the integrity of the blood-testis barrier and testosterone synthesis. Poly(I:C)-induced TNF-α production thus plays a critical role in the impairment of cell functions. In contrast, HSV60 predominantly induced the expression of type 1 interferons and antiviral proteins via the DNA sensor signaling pathway, which did not affect testicular cell functions. Accordingly, the Zika virus induced high levels of TNF-α in SC and LC and impaired their respective cellular functions, whereas Herpes simplex virus type 2 principally induced antiviral responses and did not impair such functions. These results provide insights into the mechanisms by which RNA viral infections impair testicular functions.


Subject(s)
DNA, Viral/metabolism , Leydig Cells/metabolism , RNA, Viral/metabolism , Receptors, Virus/metabolism , Sertoli Cells/metabolism , Animals , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Chlorocebus aethiops , DNA, Viral/pharmacology , Leydig Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleic Acids/metabolism , Poly I-C/metabolism , Poly I-C/pharmacology , RNA, Viral/pharmacology , Sertoli Cells/drug effects , Signal Transduction/immunology , Testis/drug effects , Testis/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL