Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 710
Filter
1.
Toxicology ; 458: 152836, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34147545

ABSTRACT

China's clean energy and resources are mainly located in the west and north while electric load center is concentrated in the middle and east. Thus, these resources and energy need to be converted into electrical energy in situ and transported to electric load center through ultra-high voltage direct current (UHVDC) transmissions. China has built 25,000 km UHVDC transmission lines of 800 kV and 1100 kV, near which the impact of electric field on health has attracted public attention. Previous studies showed that time-varying electromagnetic field exposure could disturb testosterone secretion. To study the effect of non-time-varying electric field caused by direct current transmission lines on testosterone synthesis, male ICR mice were continually (24 h/d) exposed to static electric field of 56.3 ± 1.4 kV/m. Results showed that on the 3rd day of exposure and on the 7th day after ceasing the exposure of 28 d, serum testosterone level and testicular oxidative stress indicators didn't change significantly. On the 28th day of exposure, serum testosterone levels, testicular glutathione peroxidase (GSH-Px) activity, the mRNA and protein levels of testicular StAR, PBR, CYP11A1 decreased significantly, and testicular malondialdehyde (MDA) content increased significantly. Meanwhile, electron-dense edges and vacuolation appeared in lipid droplets of Leydig cells. The gap between inner mitochondrial membrane (IMM) and outer mitochondrial membrane (OMM) enlarged, which would cause the swelling of mitochondria, the rupture and deficiency of mitochondrial membranes. Analysis showed that testicular oxidative stress could induce the damage of mitochondrial structure in Leydig cells, which would decrease the rate of cholesterol transport from cytoplasm to mitochondria. Since cholesterol is the necessary precursor of testosterone synthesis, testosterone synthesis was inhibited. The decrease of the mRNA and protein expression levels of StAR and PBR in testes could diminish the cholesterol transported from OMM to IMM. The decrease of the mRNA and protein expression levels of CYP11A1 could reduce the pregnenolone required in testosterone synthesis and inhibit testosterone synthesis consequently.


Subject(s)
Electromagnetic Fields , Leydig Cells/metabolism , Leydig Cells/radiation effects , Testosterone/biosynthesis , Animals , Antioxidants/metabolism , Cholesterol/metabolism , Cytoplasm/metabolism , Cytoplasm/radiation effects , Glutathione Peroxidase/metabolism , Leydig Cells/ultrastructure , Male , Mice , Mice, Inbred ICR , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/radiation effects , Mitochondrial Swelling/radiation effects , Oxidative Stress/radiation effects , Phosphoproteins/metabolism , Testosterone/blood , Vacuoles/radiation effects , Vacuoles/ultrastructure
2.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670702

ABSTRACT

Leydig cells contain significant amounts of constitutively produced steroidogenic acute regulatory protein (STAR; STARD1). Hormone-induced STAR plays an essential role in inducing the transfer of cholesterol into the mitochondria for hormone-dependent steroidogenesis. STAR acts at the outer mitochondrial membrane, where it interacts with a protein complex, which includes the translocator protein (TSPO). Mutations in STAR cause lipoid congenital adrenal hyperplasia (lipoid CAH), a disorder characterized by severe defects in adrenal and gonadal steroid production; in Leydig cells, the defects are seen mainly after the onset of hormone-dependent androgen formation. The function of constitutive STAR in Leydig cells is unknown. We generated STAR knockout (KO) MA-10 mouse tumor Leydig cells and showed that STAR KO cells failed to form progesterone in response to dibutyryl-cAMP and to TSPO drug ligands, but not to 22(R)-hydroxycholesterol, which is a membrane-permeable intermediate of the CYP11A1 reaction. Electron microscopy of STAR KO cells revealed that the number and size of lipid droplets were similar to those in wild-type (WT) MA-10 cells. However, the density of lipid droplets in STAR KO cells was drastically different than that seen in WT cells. We isolated the lipid droplets and analyzed their content by liquid chromatography-mass spectrometry. There was a significant increase in cholesteryl ester and phosphatidylcholine content in STAR KO cell lipid droplets, but the most abundant increase was in the amount of diacylglycerol (DAG); DAG 38:1 was the predominantly affected species. Lastly, we identified genes involved in DAG signaling and lipid metabolism which were differentially expressed between WT MA-10 and STAR KO cells. These results suggest that constitutive STAR in Leydig cells is involved in DAG accumulation in lipid droplets, in addition to cholesterol transport. The former event may affect cell functions mediated by DAG signaling.


Subject(s)
Leydig Cells/metabolism , Phosphoproteins/metabolism , Animals , Base Sequence , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Diglycerides/metabolism , Gene Deletion , Leydig Cells/ultrastructure , Ligands , Lipid Droplets/metabolism , Lipid Droplets/ultrastructure , Male , Mice, Inbred C57BL , Models, Biological , Progesterone/metabolism , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Signal Transduction , Steroids/biosynthesis
3.
Mol Cell Endocrinol ; 519: 111039, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32980418

ABSTRACT

Previous studies have demonstrated that the transplantation of alginate-poly-ʟ-lysine-alginate (APA)-encapsulated rat Leydig cells (LCs) provides a promising approach for treating testosterone deficiency (TD). Nevertheless, LCs have a limited capacity to proliferate, limiting the efficacy of LC transplantation therapy. Here, we established an efficient differentiation system to obtain functional Leydig-like cells (LLCs) from human stem Leydig cells (hSLCs). Then we injected APA-encapsulated LLCs into the abdominal cavities of castrated mice without an immunosuppressor. The APA-encapsulated cells survived and partially restored testosterone production for 90 days in vivo. More importantly, the transplantation of encapsulated LLCs ameliorated the symptoms of TD, such as fat accumulation, muscle atrophy and adipocyte accumulation in bone marrow. Overall, these results suggest that the transplantation of encapsulated LLCs is a promising new method for testosterone supplementation with potential clinical applications in TD.


Subject(s)
Cells, Immobilized/transplantation , Leydig Cells/transplantation , Testosterone/deficiency , Adipocytes/pathology , Adolescent , Adult , Aged , Alginates/chemistry , Antigens, CD/metabolism , Bone Marrow/pathology , Capsules , Castration , Cell Differentiation , Humans , Leydig Cells/ultrastructure , Male , Middle Aged , Muscular Atrophy/pathology , Polylysine/analogs & derivatives , Polylysine/chemistry , Testosterone/metabolism , Young Adult
4.
Cell Death Dis ; 11(8): 626, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796811

ABSTRACT

Abnormal lipid/lipoprotein metabolism induced by obesity may affect spermatogenesis by inhibiting testosterone synthesis in Leydig cells. It is crucial to determine which components of lipoproteins inhibit testosterone synthesis. Circulating oxidized low-density lipoprotein (oxLDL), the oxidized form of LDL, has been reported to be an independent risk factor for decreased serum testosterone levels. However, whether oxLDL has a damaging effect on Leydig cell function and the detailed mechanisms have been rarely studied. This study first showed the specific localization of oxLDL and mitochondrial structural damage in testicular Leydig cells of high-fat diet-fed mice in vivo. We also found that oxLDL reduced the mitochondrial membrane potential (MMP) by disrupting electron transport chain and inhibited testosterone synthesis-related proteins and enzymes (StAR, P450scc, and 3ß­HSD), which ultimately led to mitochondrial dysfunction and decreased testosterone synthesis in Leydig cells. Further experiments demonstrated that oxLDL promoted lipid uptake and mitochondrial dysfunction by inducing CD36 transcription. Meanwhile, oxLDL facilitated COX2 expression through the p38 MAPK signaling pathway in Leydig cells. Blockade of COX-2 attenuated the oxLDL-induced decrease in StAR and P450scc. Our clinical results clarified that the increased serum oxLDL level was associated with a decline in circulating testosterone levels. Our findings amplify the damaging effects of oxLDL and provide the first evidence that oxLDL is a novel metabolic biomarker of male-acquired hypogonadism caused by abnormal lipid metabolism.


Subject(s)
Cyclooxygenase 2/metabolism , Leydig Cells/metabolism , Lipoproteins, LDL/toxicity , Mitochondria/metabolism , Signal Transduction , Testosterone/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , 3-Hydroxysteroid Dehydrogenases/metabolism , Adult , Animals , CD36 Antigens/metabolism , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cyclooxygenase Inhibitors/pharmacology , Diet, High-Fat , Humans , Leydig Cells/drug effects , Leydig Cells/ultrastructure , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/ultrastructure , Phosphoproteins/metabolism , Semen/metabolism , Signal Transduction/drug effects , Testis/drug effects , Testis/pathology , Testis/ultrastructure , Testosterone/blood , Transcription, Genetic/drug effects , Young Adult
5.
Toxicol Lett ; 332: 213-221, 2020 Oct 10.
Article in English | MEDLINE | ID: mdl-32693021

ABSTRACT

Di-n-hexyl phthalate (DNHP) is commonly used as a plasticizer. However, whether DNHP influences Leydig cell development during puberty remains unexplored. In this study, DNHP (0, 10, 100, and 1000 mg/kg) was administered via gavage to 35-day-old male Sprague-Dawley rats for 21 days. Serum levels of testosterone, luteinizing hormone, follicle-stimulating hormone, Leydig cell number, the expression of Leydig and Sertoli cell genes and proteins were investigated. DNHP significantly increased serum testosterone levels at 10 mg/kg but lowered its level at 1000 mg/kg. DNHP significantly increased luteinizing hormone levels at 1000 mg/kg without affecting follicle-stimulating hormone levels. DNHP increased Leydig cell number at all doses but down-regulated the expression of Lhcgr, Hsd3b1, Hsd17b3, and Hsd11b1 in Leydig cell per se at 1000 mg/kg. DNHP elevated phosphorylation of ERK1/2 and GSK-3ß at 10 mg/kg but decreased SIRT1 and PGC-1α levels at 1000 mg/kg. In conclusion, DNHP exposure causes Leydig cell hyperplasia possibly via stimulating phosphorylation of ERK1/2 and GSK-3ß signaling pathways.


Subject(s)
Leydig Cells/drug effects , Phthalic Acids/toxicity , Plasticizers/toxicity , Animals , Cell Size/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 beta/drug effects , Hyperplasia , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Luteinizing Hormone/blood , MAP Kinase Signaling System/drug effects , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Sexual Maturation , Signal Transduction/drug effects , Testosterone/blood
6.
Oxid Med Cell Longev ; 2020: 4569268, 2020.
Article in English | MEDLINE | ID: mdl-32566080

ABSTRACT

Mono-(2-ethylhexyl) phthalate (MEHP) and genistein have been classified as endocrine-disrupting chemicals (EDCs) which interfere with the differentiation and development of the male reproductive system. However, how these two EDCs would affect fetal rat testis development at a low dose was rarely studied. In this study, we established the organ culture system and applied it to evaluate testicular effects following multiple EDC exposure at a low dose. 15.5 days postcoitum fetal rat testes were dissected, cultured, and exposed to vehicle (control), GEN (1 µmol/L, G), MEHP (1 µmol/L, M), or GEN (1 µmol/L)+MEHP (1 µmol/L, G+M). Testicular cell markers, testosterone concentration, redox state, testicular histology, and testicular ultrastructure were evaluated. Our results showed that a low dose of MEHP suppressed the development of Sertoli cells, Leydig cells, and gonocytes by triggering oxidative injuries, which was consistent with the ultrastructural findings. However, coadministration of genistein at a low dose could partially attenuate MEHP-induced fetal testis damage through antioxidative action. Cotreatment of genistein at a low dose may have a promising future on its protecting role for attenuating other EDC-induced reproductive disorders during early life. Based on the results, it can be speculated that dietary intake of isoflavones may make the fetal testis less susceptible to phthalate-induced injury.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Fetus/pathology , Genistein/pharmacology , Organ Culture Techniques , Testis/embryology , Testis/pathology , Animals , Antioxidants/metabolism , Basement Membrane/drug effects , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Biomarkers/metabolism , Diethylhexyl Phthalate/toxicity , Female , Gene Expression Regulation, Developmental/drug effects , Germ Cells/drug effects , Germ Cells/metabolism , Germ Cells/ultrastructure , Leydig Cells/drug effects , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Male , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Rats, Sprague-Dawley , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure , Testis/drug effects , Testis/ultrastructure , Testosterone/metabolism
7.
Eur Urol Focus ; 6(5): 1124-1129, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32563676

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), involves multiple organs. Testicular involvement is largely unknown. OBJECTIVE: To determine the pathological changes and whether SARS-CoV-2 can be detected in the testes of deceased COVID-19 patients. DESIGN, SETTING, AND PARTICIPANTS: Postmortem examination of the testes from 12 COVID-19 patients was performed using light and electron microscopy, and immunohistochemistry for lymphocytic and histiocytic markers. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the virus in testicular tissue. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Seminiferous tubular injury was assessed as none, mild, moderate, or severe according to the extent of tubular damage. Leydig cells in the interstitium were counted in ten 400× microscopy fields. RESULTS AND LIMITATIONS: Microscopically, Sertoli cells showed swelling, vacuolation and cytoplasmic rarefaction, detachment from tubular basement membranes, and loss and sloughing into lumens of the intratubular cell mass. Two, five, and four of 11 cases showed mild, moderate, and severe injury, respectively. The mean number of Leydig cells in COVID-19 testes was significantly lower than in the control group (2.2 vs 7.8, p < 0.001). In the interstitium there was edema and mild inflammatory infiltrates composed of T lymphocytes and histiocytes. Transmission EM did not identify viral particles in three cases. RT-PCR detected the virus in one of 12 cases. CONCLUSIONS: Testes from COVID-19 patients exhibited significant seminiferous tubular injury, reduced Leydig cells, and mild lymphocytic inflammation. We found no evidence of SARS-CoV-2 virus in the testes in the majority (90%) of the cases by RT-PCR, and in none by electron microscopy. These findings can provide evidence-based guidance for sperm donation and inform management strategies to mitigate the risk of testicular injury during the COVID-19 disease course. PATIENT SUMMARY: We examined the testes of deceased COVID-19 patients. We found significant damage to the testicular parenchyma. However, virus was not detected in testes in the majority of cases.


Subject(s)
Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Seminiferous Tubules/pathology , Testis/pathology , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2 , Betacoronavirus , COVID-19 , Cell Count , Coronavirus Infections/metabolism , Coronavirus Infections/physiopathology , Humans , Inflammation , Leydig Cells/pathology , Leydig Cells/ultrastructure , Male , Microscopy, Electron , Middle Aged , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Seminiferous Tubules/ultrastructure , Sertoli Cells/pathology , Sertoli Cells/ultrastructure , Spermatogenesis/physiology , Testis/metabolism , Testis/ultrastructure , Testis/virology
8.
Int J Mol Sci ; 21(10)2020 May 21.
Article in English | MEDLINE | ID: mdl-32455738

ABSTRACT

Although epidemiological studies from the last years report an increase in the incidences of Leydig cell tumors (previously thought to be a rare disease), the biochemical characteristics of that tumor important for understanding its etiology, diagnosis, and therapy still remains not completely characterized. Our prior studies reported G-protein coupled estrogen receptor signaling and estrogen level disturbances in Leydig cell tumors. In addition, we found that expressions of multi-level-acting lipid balance- and steroidogenesis-controlling proteins including peroxisome proliferator-activated receptor are altered in this tumor. In order to get deeper into the other molecular mechanisms that regulate lipid homeostasis in the Leydig cell tumor, here we investigate the presence and expression of newly-described hormones responsible for lipid homeostasis balancing (leptin and adiponectin), together with expression of estrogen synthase (aromatase). Samples of Leydig cell tumors (n = 20) were obtained from patients (31-45 years old) and used for light and transmission electron microscopic, western blotting, and immunohistochemical analyses. In addition, body mass index (BMI) was calculated. In tumor mass, abundant lipid accumulation in Leydig cells and various alterations of Leydig cell shape, as well as the presence of adipocyte-like cells, were observed. Marked lipid content and various lipid droplet size, especially in obese patients, may indicate alterations in lipid homeostasis, lipid processing, and steroidogenic organelle function in response to interstitial tissue pathological changes. We revealed significantly increased expression of leptin, adiponectin and their receptors, as well as aromatase in Leydig cell tumors in comparison to control. The majority of patients (n = 13) were overweight as indicated by their BMI. Moreover, a significant increase in expression of phospholipase C (PLC), and kinases Raf, ERK which are part of adipokine transductional pathways, was demonstrated. These data expand our previous findings suggesting that in human Leydig cell tumors, estrogen level and signaling, together with lipid status, are related to each other. Increased BMI may contribute to certain biochemical characteristics and function of the Leydig cell in infertile patients with a tumor. In addition, altered adipokine-estrogen microenvironment can have an effect on proliferation, growth, and metastasis of tumor cells. We report here various targets (receptors, enzymes, hormones) controlling lipid balance and estrogen action in Leydig cell tumors indicating their possible usefulness for diagnostics and therapy.


Subject(s)
Adiponectin/metabolism , Aromatase/metabolism , Carcinogenesis/metabolism , Leptin/metabolism , Leydig Cell Tumor/metabolism , Adult , Carcinogenesis/ultrastructure , Humans , Leydig Cell Tumor/ultrastructure , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Lipid Droplets/metabolism , Male , Signal Transduction
9.
Acta Histochem ; 122(3): 151526, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32094002

ABSTRACT

Communication in biological systems involves diverse-types of cell-cell interaction including cross-talk between receptors expressed by the target cells. Recently, novel sort of estrogen receptors (G protein - coupled estrogen receptor; GPER and estrogen-related receptor; ERR) that signal directly via estrogen binding and/or via mutual interaction-regulated estrogen signaling were reported in various organs including testis. Peroxisome proliferator - activated receptor (PPAR) is responsible for maintaining of lipid homeostasis that is critical for sex steroid production in the testis. Here, we investigated the role of interaction between GPER, ERRß and PPARγ in steroidogenic Leydig cells of immature boar testis. Testicular fragments cultured ex vivo were treated with GPER or PPARγ antagonists. Then, cell ultrastructure, expression and localization of GPER, ERRß, PPARγ together with the molecular receptor mechanism, through cyclic AMP and Raf/Ras/extracellular signal activated kinases (ERK), in the control of cholesterol concentration and estrogen production by Leydig cells were studied. In the ultrastructure of antagonist-treated Leydig cells, mitochondria were not branched and not bifurcated as they were found in control. Additionally, in PPARγ-blocked Leydig cells changes in the number of lipid droplets were revealed. Independent of used antagonist, western blot revealed decreased co-expression of GPER, ERRß, PPARγ with exception of increased expression of ERRß after PPARγ blockage. Immunohistochemistry confirmed presence of all receptors partially located in the nucleus or cytoplasm of Leydig cells of both control and treated testes. Changes in receptor expression, decreased cholesterol and increased estradiol tissue concentrations occurred through decreased cAMP level (with exception after GPER blockage) as well as Raf/Ras/ERK pathway expression. These all findings indicate that GPER-ERRß-PPARγ interaction exists in immature boar testis and regulates Leydig cell function. Further detailed studies and considerations on GPER-ERRß-PPARγ as possible diagnosis/therapy target in disturbances of testis steroidogenic function are needed.


Subject(s)
Leydig Cells/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Estrogen/metabolism , Testis/metabolism , Animals , Cell Nucleus/metabolism , Cholesterol/metabolism , Cyclic AMP/metabolism , Cytoplasm/metabolism , Estrogen Receptor beta/metabolism , Estrogens/biosynthesis , Leydig Cells/ultrastructure , MAP Kinase Signaling System/drug effects , Male , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Receptors, G-Protein-Coupled/metabolism , Swine , Testis/growth & development
10.
Proc Natl Acad Sci U S A ; 116(46): 23274-23283, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31591190

ABSTRACT

Reduced serum testosterone (T), or hypogonadism, affects millions of men and is associated with many pathologies, including infertility, cardiovascular diseases, metabolic syndrome, and decreased libido and sexual function. Administering T-replacement therapy (TRT) reverses many of the symptoms associated with low T levels. However, TRT is linked to side effects such as infertility and increased risk of prostate cancer and cardiovascular diseases. Thus, there is a need to obtain T-producing cells that could be used to treat hypogonadism via transplantation and reestablishment of T-producing cell lineages in the body. T is synthesized by Leydig cells (LCs), proposed to derive from mesenchymal cells of mesonephric origin. Although mesenchymal cells have been successfully induced into LCs, the limited source and possible trauma to donors hinders their application to clinical therapies. Alternatively, human induced pluripotent stem cells (hiPSCs), which are expandable in culture and have the potential to differentiate into all somatic cell types, have become the emerging source of autologous cell therapies. We have successfully induced the differentiation of hiPSCs into either human Leydig-like (hLLCs) or adrenal-like cells (hALCs) using chemically defined culture conditions. Factors critical for the development of LCs were added to both culture systems. hLLCs expressed all steroidogenic genes and proteins important for T biosynthesis, synthesized T rather than cortisol, secreted steroid hormones in response to dibutyryl-cAMP and 22(R)-hydroxycholesterol, and displayed ultrastructural features resembling LCs. By contrast, hALCs synthesized cortisol rather than T. The success in generating hiPSC-derived hLLCs with broad human LC (hLC) features supports the potential for hiPSC-based hLC regeneration.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Induced Pluripotent Stem Cells , Leydig Cells/enzymology , Testosterone/metabolism , Gene Expression , Humans , Leydig Cells/ultrastructure , Male , Transcriptome
11.
J Cell Mol Med ; 23(10): 6859-6871, 2019 10.
Article in English | MEDLINE | ID: mdl-31373170

ABSTRACT

OBJECTIVE: The high-fat diet (HFD)-induced obesity is responsible for the testosterone deficiency (TD). However, the mechanism remains unknown. Mitochondrial homeostasis is proved to be important for maintaining the function of steroidogenic acute regulatory protein (StAR), the first rate-limiting enzyme in testosterone synthesis. As the key regulator of mitochondrial membrane permeability, cyclophilin D (CypD) plays a crucial role in maintaining mitochondrial function. In this study, we sought to elucidate the role of CypD in the expression of StAR affected by HFD. METHODS: To analyse the influence of CypD on StAR in vivo and in vitro, mouse models of HFD, CypD overexpression and CypD knockout (Ppif-/- ) as well as Leydig cells treated with palmitic acid (PA) and CypD overexpression plasmids were examined with an array of metabolic, mitochondrial function and molecular assays. RESULTS: Compared with the normal diet mice, consistent with reduced testosterone in testes, the expressions of StAR in both mRNA and protein levels in HFD mice were down-regulated, while expressions of CypD were up-regulated. High-fat intake impaired mitochondrial function with the decrease in StAR in Leydig cells. Overexpression of CypD inhibited StAR expressions in vivo and in vitro. Compared with C57BL/6 mice with HFD, expressions of StAR were improved in Ppif-/- mice with HFD. CONCLUSIONS: Mitochondrial CypD involved in the inhibitory effect of HFD on StAR expression in testes.


Subject(s)
Diet, High-Fat , Peptidyl-Prolyl Isomerase F/metabolism , Phosphoproteins/metabolism , Animals , Down-Regulation/genetics , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Lipid Metabolism , Lipids/toxicity , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/ultrastructure , Phosphoproteins/genetics , Up-Regulation/genetics
12.
Theriogenology ; 138: 111-120, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31325741

ABSTRACT

Multilamellar bodies (MLBs) are produced and secreted by many cell types. In this study, we report the existence and ultrastructure of MLBs that are produced by Leydig cells and identification of telocytes in the testicular interstitium of naked mole rat. This study was performed on both breeder and non-breeder male naked mole rats using light microscopy, transmission electron microscopy, and morphometric approaches. In the testicular interstitium, the most prominent cells were Leydig cells, which contained numerous lipid droplets (LDs) in the cytoplasm. We found that MLBs were associated with the LDs of Leydig cells and were secreted into the extracellular or interstitial environment via exocytosis. After their release from Leydig cells, MLBs localized to the space between Leydig cells near blood vessels and attached to telocytes. We also identified telocytes in the testicular interstitium, and their cellular extensions were distributed throughout the interstitium. MLBs were aligned along the cellular extensions of telocytes, and membrane-to-membrane contact was observed between the cellular extensions of telocytes and MLBs, suggesting that telocytes may play a role in the transport of MLBs within the interstitial space. No ultrastructural differences were found in Leydig cells, telocytes, or MLBs between breeder and non-breeder testes. However, morphometric analysis revealed a significant difference in the number of MLBs between the breeder and non-breeder animals. Furthermore, both selective autophagy of LDs and non-selective autophagy were observed in Leydig cells. Typical features of macrolipophagy were also observed, as a few LDs were entirely enclosed by a limiting membrane. Remarkably, autophagy may be a key factor in the biogenesis of MLBs and steroid hormone production. The appearance of MLBs in the testicular interstitium of naked mole rats could thus be related to lipid storage and trafficking.


Subject(s)
Inclusion Bodies/ultrastructure , Leydig Cells/cytology , Leydig Cells/ultrastructure , Mole Rats , Telocytes/cytology , Testis , Animals , Autophagy/physiology , Leydig Cells/physiology , Male , Microscopy , Microscopy, Electron, Transmission , Telocytes/ultrastructure , Testis/cytology , Testis/ultrastructure
13.
Acta Histochem ; 121(5): 611-618, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31126612

ABSTRACT

In this study mouse Leydig cell (MA-10) were treated with G-protein coupled membrane estrogen receptor antagonist (G-15; 10 nM). Cells were analyzed by Western blotting for expression of estrogen-related receptors (ERRα, ß and γ), steroidogenic markers (lutropin receptor; LHR and 3ß-hydroxysteroid dehydrogenase; 3ß-HSD) and lipid droplet markers (perilipin; PLIN and microtubule-associated protein 1 A/1B-light chain 3; LC3). Concomitantly, microscopic analyses by light microscope (immunofluorescent staining for lipid droplets, PLIN and LC3) as well as by electron microscope (for lipid droplet ultrastructure) were utilized. For analysis of cholesterol content, cAMP level and progesterone secretion, G-15, estrogen receptor (ER) antagonist (ICI 182,780; 10 µM), 17ß-estradiol (10 mM) and, bisphenol A (BPA; 10 nM) were used alone or in combinations. We revealed no changes in ERRs expression but alterations in ERRß and γ localization in G-15-treated cells when compared to control. Partial translocation of ERRß and γ from the cell nucleus to cytoplasm was observed. Decreased expression of LHR, 3ß-HSD, PLIN and LC3 was detected. Moreover, in treated cells large lipid droplets and differences in their distribution were found. Very strong signal of co-localization for PLIN and LC3 was found in treated cells when compared to control. In ultrastructure of treated cells, degenerating lipid droplets and double membrane indicating on presence of lipophagosome were observed. We found, that only (i) BPA and G-15 did not effect on cholesterol content, (ii) BPA, G-15 and ICI did not effect on cAMP level and (iii) BPA, ICI alone and in combination, and BPA with G-15 did not modulate progesterone secretion. These findings showed complex and diverse estrogen effects on mouse Leydig cells at various steps of steroid hormone production (cholesterol storage, release and processing). Lipid homeostasis and metabolism in these cells were affected by endogenous and exogenous estrogen, interactions of receptors (GPER, ER and ERR) and GPER and ER antagonists.


Subject(s)
Estrogens/physiology , Leydig Cells/metabolism , Lipid Metabolism/physiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Estrogens/pharmacology , Leydig Cells/drug effects , Leydig Cells/ultrastructure , Lipid Droplets/ultrastructure , Male , Mice , ERRalpha Estrogen-Related Receptor
14.
Bull Exp Biol Med ; 166(6): 802-805, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31028585

ABSTRACT

Sialadenectomy in young rats modifies the development of the spermatogenic and steroidogenic functions of the testes. Sialadenectomy causes ultrastructural changes in spermatogenic cells, sustentocytes, and Leydig cells that disappear by week 8 of the experiment due to realization of compensatory and adaptive mechanisms. The effects of endocrine factors of the greater salivary glands on the spermatogenic cells are realized directly and indirectly via interstitial endocrinocytes and sustentocytes.


Subject(s)
Leydig Cells/ultrastructure , Salivary Glands/surgery , Seminiferous Epithelium/ultrastructure , Sertoli Cells/ultrastructure , Adaptation, Physiological , Animals , Animals, Outbred Strains , Male , Rats , Salivary Glands/physiology , Spermatogenesis/physiology , Time Factors
15.
Reprod Biol Endocrinol ; 17(1): 19, 2019 Feb 09.
Article in English | MEDLINE | ID: mdl-30738428

ABSTRACT

BACKGROUND: Steroidogenesis is an indispensable process that is indirectly associated with spermatogenesis in the Leydig cell (LC) to utilize the lipid droplets (LDs) that are critical to maintaining normal testosterone synthesis. The regulation of LD mobilization, known as lipophagy, in the LC is still largely unknown. METHOD: In the present study, the LC of the Chinese soft-shelled turtle was investigated to identify the steroidogenic activity and lipophagy during the annual reproductive cycle by light microscopy, immunohistochemistry (IHC), immunofluorescence (IF), and transmission electron microscopy (TEM). RESULTS: The LC showed a dynamic steroidogenic function with strong activity of 3ß-HSD, vimentin and tubular ER during hibernation by IHC and TEM. The tubulo-vesicular ER had a weak immunopositive reaction for 3ß-HSD in the LC during reproductive phase, suggesting persistent steroidogenic activity. ORO staining and TEM demonstrated that a larger number of LDs had accumulated in the LC during hibernation than in the reproductive phase. These LDs existed in close association with mitochondria and lysosomes by being dynamically surrounded by intermediate filaments to facilitate LD utilization. Lysosomes were found directly attached to large LDs, forming an autophagic tube and engulfing LDs, suggesting that micro-lipophagy occurs during hibernation. Furthermore, the IHC of ATG7 (Autophagy Related Gene 7) and the IF of the LC3 (Microtubule-associated protein light chain 3), p62 (Sequestosome-1 (SQSTM1) and LAMP1(Lysosomal-associated membrane protein 1) results demonstrated strong expression, and further confirmation by TEM showed the existence of an autophagosome and an autolysosome and their fusion during the hibernation season. CONCLUSION: In conclusion, the present study provides clear evidence of LD consumption in the LC by lipophagy, lysosome and mitochondria during the hibernation period, which is a key aspect of steroidogenesis in the Chinese soft-shelled turtle.


Subject(s)
Autophagy/physiology , Leydig Cells/metabolism , Lipid Metabolism , Steroids/metabolism , Turtles/metabolism , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Hibernation/physiology , Leydig Cells/ultrastructure , Lipid Droplets/metabolism , Lipid Droplets/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondria/ultrastructure , Reproduction/physiology , Turtles/physiology
16.
Bull Exp Biol Med ; 166(2): 279-282, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30488201

ABSTRACT

We studied immunohistochemical and morphometric characteristics of the spermatogenic epithelium in rats against the background of peroral administration of nanoparticles containing titanium dioxide. Substantial degenerative changes in the spermatogenic epithelium were revealed: thinning, disorganization of layers, and detachment of sperm cells from the basement membrane. Immunohistochemical analysis revealed reduced proliferative activity and differentiation potential of epithelial cells, which was confirmed by changes in the expression of Ki-67 and c-kit markers. Our data attest to unfavorable effects of titanium dioxide nanoparticles on the structural and functional characteristics of the reproductive system in male rats leading to spermatogenesis disturbances.


Subject(s)
Leydig Cells/drug effects , Nanoparticles/administration & dosage , Seminiferous Tubules/drug effects , Sertoli Cells/drug effects , Spermatogenesis/drug effects , Titanium/administration & dosage , Administration, Oral , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Ki-67 Antigen/metabolism , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Male , Nanoparticles/chemistry , Rats , Rats, Wistar , Seminiferous Tubules/metabolism , Seminiferous Tubules/ultrastructure , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure , Sperm Count , Spermatozoa/cytology , Spermatozoa/drug effects
17.
Sci Rep ; 8(1): 14780, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30283023

ABSTRACT

Telocytes are a recently described stromal cell type widely distributed in various organs including the female and male reproductive systems. This study was aimed to investigate for the first time the existence, distribution and characteristics of telocytes in normal human testis by an integrated morphological approach (immunohistochemistry, immunofluorescence and transmission electron microscopy). We found that telocytes displaying typical long and moniliform prolongations and coexpressing CD34 and PDGFRα formed networks in the outer layer of peritubular tissue and around Leydig cells and vessels in the intertubular stroma. Testicular telocytes were immunophenotypically negative for CD31, c-kit/CD117 as well as α-SMA, thus making them clearly distinguishable from myoid cells/myofibroblasts located in the inner layer of peritubular tissue. Transmission electron microscopy confirmed the presence of cells ultrastructurally identifiable as telocytes (i.e. cells with telopodes alternating podomers and podoms) in the aforementioned locations. Intercellular contacts between neighboring telocytes and telopodes were observed throughout the testicular stromal compartment. Telopodes intimately surrounded and often established close contacts with peritubular myoid cells/myofibroblasts, Leydig cells and vessels. Extracellular vesicles were also frequently detected near telopodes. In summary, we demonstrated that telocytes are a previously neglected stromal component of human testis with potential implications in tissue homeostasis deserving further investigation.


Subject(s)
Leydig Cells/ultrastructure , Telocytes/ultrastructure , Telopodes/ultrastructure , Testis/ultrastructure , Antigens, CD34/genetics , Connective Tissue/metabolism , Connective Tissue/ultrastructure , Gene Expression Regulation , Humans , Immunohistochemistry , Immunophenotyping , Leydig Cells/metabolism , Male , Microscopy, Electron, Transmission , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stromal Cells/metabolism , Stromal Cells/ultrastructure , Telocytes/metabolism , Telopodes/metabolism , Testis/metabolism
18.
Int J Nanomedicine ; 13: 5909-5924, 2018.
Article in English | MEDLINE | ID: mdl-30319256

ABSTRACT

BACKGROUND: Nanoparticulate titanium dioxide (nano-TiO2) enters the body through various routes and causes organ damage. Exposure to nano-TiO2 is reported to cause testicular injury in mice or rats and decrease testosterone synthesis, sperm number, and motility. Importantly, nano-TiO2 suppresses testosterone production by Leydig cells (LCs) and impairs the reproductive capacity of animals. METHODS: In an attempt to establish the molecular mechanisms underlying the inhibitory effect of nano-TiO2 on testosterone synthesis, primary cultured rat LCs were exposed to varying concentrations of nano-TiO2 (0, 10, 20, and 40 µg/mL) for 24 hours, and alterations in cell viability, cell injury, testosterone production, testosterone-related factors (StAR, 3ßHSD, P450scc, SR-BI, and DAX1), and signaling molecules (ERK1/2, PKA, and PKC) were investigated. RESULTS: The data show that nano-TiO2 crosses the membrane into the cytoplasm or nucleus, triggering cellular vacuolization and nuclear condensation. LC viability decreased in a time-dependent manner at the same nano-TiO2 concentration, nano-TiO2 treatment (10, 20, and 40 µg/mL) decreased MMP (36.13%, 45.26%, and 79.63%), testosterone levels (11.40% and 44.93%), StAR (14.7%, 44.11%, and 72.05%), 3ßHSD (26.56%, 50%, and 79.69%), pERK1/2 (27.83%, 63.61%, and 78.89%), PKA (47.26%, 70.54%, and 85.61%), PKC (30%, 50%, and 71%), SR-BI (16.41%, 41.79%, and 67.16%), and P450scc (39.41%, 55.26%, and 86.84%), and upregulated DAX1 (1.31-, 1.63-, and 3.18-fold) in primary cultured rat LCs. CONCLUSION: Our collective findings indicated that nano-TiO2-mediated suppression of testosterone in LCs was associated with regulation of ERK1/2-PKA-PKC signaling pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Leydig Cells/metabolism , MAP Kinase Signaling System/drug effects , Nanoparticles/chemistry , Protein Kinase C/metabolism , Testosterone/biosynthesis , Titanium/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Endocytosis/drug effects , Hydrodynamics , Leydig Cells/drug effects , Leydig Cells/ultrastructure , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Models, Biological , Nanoparticles/ultrastructure , Rats , Testosterone/metabolism , X-Ray Diffraction
19.
Bull Exp Biol Med ; 165(2): 280-283, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29931628

ABSTRACT

The major salivary glands of rats release into the saliva and blood a wide spectrum of bioactive substances, essential for many organs, including the testes. Sialoadenectomy leads to the development of degenerative changes in the cells of the twisted testicular tubules. However, the effects of bioactive factors released by the major salivary glands on the morphology and function of Leydig cells remain little studied. Sialoadenectomy in adult rats led (in 1-4 weeks) to a decrease in the nuclear and cytoplasmatic areas of Leydig cells, violation of the plasmalemma integrity, dilatation of perinuclear space and agranular endoplasmatic reticulum vesicles, and to destruction of the mitochondria. Ultrastructural changes caused by sialoadenectomy completely resolved by week 6 of the experiment at the expense of compensatory activation of the synthesis of the major salivary gland factors by other sources in the organism of rats.


Subject(s)
Leydig Cells/ultrastructure , Salivary Glands/surgery , Sexual Maturation/physiology , Testis/ultrastructure , Age Factors , Animals , Animals, Outbred Strains , Endocrine Surgical Procedures , Leydig Cells/cytology , Male , Rats , Saliva/metabolism , Salivary Glands/pathology , Testis/cytology , Testis/metabolism
20.
Cell Tissue Res ; 374(2): 389-412, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29876633

ABSTRACT

In this study, G-coupled estrogen receptor (GPER) was inactivated, by treatment with antagonist (G-15), in testes of C57BL/6 mice: immature (3 weeks old), mature (3 months old) and aged (1.5 years old) (50 µg/kg bw), as well as MA-10 mouse Leydig cells (10 nM/24 h) alone or in combination with 17ß-estradiol or antiestrogen (ICI 182,780). In G-15-treated mice, overgrowth of interstitial tissue was found in both mature and aged testes. Depending on age, differences in structure and distribution of various Leydig cell organelles were observed. Concomitantly, modulation of activity of the mitochondria and tubulin microfibers was revealed. Diverse and complex GPER regulation at the mRNA level and protein of estrogen signaling molecules (estrogen receptor α and ß; ERα, ERß and cytochrome P450 aromatase; P450arom) in G-15 Leydig cells was found in relation to age and the experimental system utilized (in vivo and in vitro). Changes in expression patterns of ERs and P450arom, as well as steroid secretion, reflected Leydig cell heterogeneity to estrogen regulation throughout male life including cell physiological status.We show, for the first time, GPER with ERs and P450arom work in tandem to maintain Leydig cell architecture and supervise its steroidogenic function by estrogen during male life. Full set of estrogen signaling molecules, with involvement of GPER, is crucial for proper Leydig cell function where each molecule acts in a specific and/or complementary manner. Further understanding of the mechanisms by which GPER controls Leydig cells with special regard to male age, cell of origin and experimental system used is critical for predicting and preventing testis steroidogenic disorders based on perturbations in estrogen signaling.


Subject(s)
Leydig Cells/cytology , Leydig Cells/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Cell Shape , Cytoskeleton/metabolism , Leydig Cells/ultrastructure , Male , Mice, Inbred C57BL , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Steroids/metabolism , Testis/cytology , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...