Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.231
Filter
1.
Anat Histol Embryol ; 53(4): e13062, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38837469

ABSTRACT

Although the order Rodentia does not present a high risk of extinction compared to mammals as a whole, several families demonstrate high levels of threat and/or data deficiency, therefore highlighting the need for targeted research and the application of ecological and reproductive data to the development of conservation actions. The order Rodentia, the largest among mammals, includes 9 families, and the family Cricetidae is the most diverse of the Brazilian rodents. In Brazil, 12 of the 16 genera of Oecomys are found. Oecomys bicolor is known in Brazil as the 'arboreal rat' and is, found in dry, deciduous and tropical forests. The mean body weight of Oecomys bicolor was 35.8 g and the gonadal, tubular and epithelial somatic indexes were, 0.53%, 0.47% and 0.37%, respectively. Seminiferous tubules volume density was 89.72% and the mitotic and meiotic indexes corresponded to 8.59 and 2.45 cells, respectively, and the yield of spermatogenesis was 23.83 cells. The intertubular compartment represented 10.28% of the testis parenchyma and around 5% of the interstitial space was occupied by Leydig cells, whose number per gram of testis was 11.10 × 107 cells. By evaluating the biometric and histomorphometric characteristics of the testis, there is evidence that this species has a high investment in reproduction. Due to the high contribution of the seminiferous epithelium and the intertubular compartment in this species, compared to the others of the same family, it is possible to infer that the species Oecomys bicolor has a promiscuous reproductive behaviour.


Subject(s)
Arvicolinae , Leydig Cells , Spermatogenesis , Testis , Animals , Spermatogenesis/physiology , Male , Testis/anatomy & histology , Testis/physiology , Leydig Cells/cytology , Leydig Cells/physiology , Arvicolinae/anatomy & histology , Arvicolinae/physiology , Seminiferous Tubules/anatomy & histology , Brazil
2.
Cell Commun Signal ; 22(1): 330, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879537

ABSTRACT

Sex-specific gonadal differentiation is directed by complex signalling promoting development in either male or female direction, while simultaneously inhibiting the opposite pathway. In mice, the WNT/ß-catenin pathway promotes ovarian development and the importance of actively inhibiting this pathway to ensure normal testis development has been recognised. However, the implications of alterations in the tightly regulated WNT/ß-catenin signalling during human fetal gonad development has not yet been examined in detail. Thus, the aim of this study was to examine the consequences of dysregulating the WNT/ß-catenin signalling pathway in the supporting cell lineage during sex-specific human fetal gonad development using an established and extensively validated ex vivo culture model. Inhibition of WNT/ß-catenin signalling in human fetal ovary cultures resulted in only minor effects, including reduced secretion of RSPO1 and reduced cell proliferation although this was not consistently found in all treatment groups. In contrast, promotion of WNT/ß-catenin signalling in testes severely affected development and function. This included disrupted seminiferous cord structures, reduced cell proliferation, reduced expression of SOX9/AMH, reduced secretion of Inhibin B and AMH as well as loss of the germ cell population. Additionally, Leydig cell function was markedly impaired with reduced secretion of testosterone, androstenedione and INSL3. Together, this study suggests that dysregulated WNT/ß-catenin signalling during human fetal gonad development severely impairs testicular development and function. Importantly, our study highlights the notion that sufficient inhibition of the opposite pathway during sex-specific gonadal differentiation is essential to ensure normal development and function also applies to human fetal gonads.


Subject(s)
Testis , Wnt Signaling Pathway , Humans , Male , Testis/metabolism , Testis/embryology , Female , Sex Differentiation/genetics , Fetus/metabolism , Cell Differentiation , Cell Proliferation , beta Catenin/metabolism , Leydig Cells/metabolism , Leydig Cells/cytology , Ovary/metabolism , Ovary/embryology
3.
J Agric Food Chem ; 72(18): 10616-10626, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38656193

ABSTRACT

Deoxynivalenol (DON) is a common food contaminant that can impair male reproductive function. This study investigated the effects and mechanisms of DON exposure on progenitor Leydig cell (PLC) development in prepubertal male rats. Rats were orally administrated DON (0-4 mg/kg) from postnatal days 21-28. DON increased PLC proliferation but inhibited PLC maturation and function, including reducing testosterone levels and downregulating biomarkers like HSD11B1 and INSL3 at ≥2 mg/kg. DON also stimulated mitochondrial fission via upregulating DRP1 and FIS1 protein levels and increased oxidative stress by reducing antioxidant capacity (including NRF2, SOD1, SOD2, and CAT) in PLCs in vivo. In vitro, DON (2-4 µM) inhibited PLC androgen biosynthesis, increased reactive oxygen species production and protein levels of DRP1, FIS1, MFF, and pAMPK, decreased mitochondrial membrane potential and MFN1 protein levels, and caused mitochondrial fragmentation. The mitochondrial fission inhibitor mdivi-1 attenuated DON-induced impairments in PLCs. DON inhibited PLC steroidogenesis, increased oxidative stress, perturbed mitochondrial homeostasis, and impaired maturation. In conclusion, DON disrupts PLC development in prepubertal rats by stimulating mitochondrial fission.


Subject(s)
Leydig Cells , Mitochondria , Mitochondrial Dynamics , Oxidative Stress , Rats, Sprague-Dawley , Trichothecenes , Animals , Male , Mitochondrial Dynamics/drug effects , Rats , Leydig Cells/drug effects , Leydig Cells/metabolism , Leydig Cells/cytology , Trichothecenes/toxicity , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Testosterone/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Humans , Dynamins/metabolism , Dynamins/genetics , Membrane Potential, Mitochondrial/drug effects
4.
Nat Commun ; 13(1): 4020, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35821241

ABSTRACT

Male reproductive system ageing is closely associated with deficiency in testosterone production due to loss of functional Leydig cells, which are differentiated from stem Leydig cells (SLCs). However, the relationship between SLC differentiation and ageing remains unknown. In addition, active lipid metabolism during SLC differentiation in the reproductive system requires transportation and processing of substrates among multiple organelles, e.g., mitochondria and endoplasmic reticulum (ER), highlighting the importance of interorganelle contact. Here, we show that SLC differentiation potential declines with disordered intracellular homeostasis during SLC senescence. Mechanistically, loss of the intermediate filament Nestin results in lower differentiation capacity by separating mitochondria-ER contacts (MERCs) during SLC senescence. Furthermore, pharmacological intervention by melatonin restores Nestin-dependent MERCs, reverses SLC differentiation capacity and alleviates male reproductive system ageing. These findings not only explain SLC senescence from a cytoskeleton-dependent MERCs regulation mechanism, but also suggest a promising therapy targeting SLC differentiation for age-related reproductive system diseases.


Subject(s)
Endoplasmic Reticulum , Leydig Cells , Mitochondria , Aging/metabolism , Cell Differentiation/physiology , Endoplasmic Reticulum/metabolism , Humans , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Mitochondria/metabolism , Nestin/metabolism
5.
Food Chem Toxicol ; 160: 112805, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34990787

ABSTRACT

Bisphenol S (BPS), an increasingly used alternative to bisphenol A, has been linked to testosterone deficiency and male reproductive dysfunction in laboratory animals. This study aimed to examine the cytotoxicity of BPS exposure to Leydig cells and to investigate its possible mechanisms. After treatment with BPS (100, 200 and 400 µM) for 48 h in vitro, TM3 mouse Leydig cells exhibited a dose-dependent decrease in the viability. Furthermore, BPS challenge triggered oxidative stress manifested by compromised activities of superoxide dismutase and catalase with exaggerated formation of reactive oxygen species. Especially, BPS exposure resulted in augmented mitochondrial permeability transition pore opening, dissipated mitochondrial membrane potential and reduced ATP generation, along with an altered energy metabolism. Moreover, BPS stimulation enhanced BAX expression and caspase-3 activity and inhibited BCL-2 expression. In addition, BPS-treated TM3 cells showed an accumulation of autophagic vacuoles, together with increased Beclin1 and P62 expression and elevated LC3B-II/LC3B-I ratio. These results demonstrated that in vitro exposure to BPS exerted cytotoxicity to TM3 Leydig cells through inducing oxidative stress, mitochondrial impairment, autophagic disturbance and apoptosis.


Subject(s)
Benzhydryl Compounds/toxicity , Leydig Cells/drug effects , Phenols/toxicity , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
6.
Gene ; 812: 146112, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-34896518

ABSTRACT

The Glial cell-derived neurotrophic factor (Gdnf) and testosterone induce the spermatogonial stem cells (SSCs) self-renewal and spermatogenesis, respectively. In present study the stimulating role of testosterone on Sertoli cells to produce Gdnf, and the possible effect of Gdnf on Gfrα1 and c-RET expressions were investigated. The TM4 cells (line Sertoli cells) were co-cultured with [0.1, 0.2 and 0.4 (ng/ml)] of exogenous and TM3 (line Leydig cells)-produced testosterones, and consequently the TM4-produced Gdnf concentration was evaluated. Next, the SSCs were co-cultured with the TM-4 derived media (endogenous Gdnf) and exogenous Gdnf [0.1, 0.2, and 0.4 ng/ml)]. The 0.1 and 0.2 ng/ml endogenous and 3 concentrations of exogenous testosterone up-regulated the Gdnf expression versus non-treated Sertoli cells. The TM4-produced and exogenous Gdnfs, in all concentrations, up-regulated the receptors expression. In conclusion, the testosterone, solely, stimulates the Gdnf synthesis and the Gdnf, individually, amplifies its receptor's expression at mRNA and protein levels.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/metabolism , Leydig Cells/cytology , Sertoli Cells/cytology , Testosterone/metabolism , Animals , Cell Line , Cell Survival/drug effects , Coculture Techniques , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Leydig Cells/metabolism , Male , Mice , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testosterone/pharmacology , Up-Regulation
7.
Oxid Med Cell Longev ; 2021: 3575016, 2021.
Article in English | MEDLINE | ID: mdl-34887997

ABSTRACT

Leydig cell injury has been described as a primary driver of testicular dysfunction and is affected by oxidative stress. Dioscorea polystachya (Chinese yam) is used to improve testicular dysfunction in clinical and pharmacological research via its antioxidative activity, but the mechanisms underlying the beneficial effect of Chinese yam on testicular dysfunction and its suppression of Leydig cell oxidative damage remain unclear. In this study, we obtained a Chinese yam protein (DP1) and explored its effectiveness and possible mechanism in improving testicular dysfunction in vivo and in vitro. We established a testicular dysfunction model in rats using hydrocortisone (HCT). DP1 increased body weight and organ index, improved the deterioration in testicular morphology (including increasing the diameter of seminiferous tubules and thickness of germinal cell layers, inhibiting testicular cell apoptosis by increasing the Bcl-2/Bax ratio, and impeding collagen leakage by downregulating TGF-ß1 and p-SMAD2/3 expression), and restored the testosterone content. In addition, DP1 enhanced the number of Leydig cells in rats and H2O2-induced TM3 Leydig cells, and the effect of DP1 on the apoptosis, fibrosis, and testosterone content of TM3 cells was similar to that observed in vivo. These changes were dependent on the regulation of oxidative stress, including significantly reduced intracellular 8-hydroxy-2-deoxyguanosine levels, enhanced superoxide dismutase activities, and decreased superoxide anion levels, which were confirmed via a superoxide overexpression system. Furthermore, we observed that DP1 promoted Nrf2 nuclear import and upregulated antioxidant factor expression in vivo and in vitro. However, Nrf2 silencing eliminated the ability of DP1 to increase the Bcl-2/Bax ratio, reduce the expression levels of TGF-ß1 and p-SMAD2/3, and increase testosterone contents in H2O2-induced TM3 cells. In conclusion, DP1 reversed the HCT-induced testicular apoptosis and fibrosis and decreased testosterone contents by alleviating Leydig cell oxidative damage via upregulation of the Nrf2 pathway.


Subject(s)
Dioscorea/metabolism , Hydrocortisone/pharmacology , NF-E2-Related Factor 2/metabolism , Plant Proteins/pharmacology , Up-Regulation/drug effects , Animals , Apoptosis/drug effects , Leydig Cells/cytology , Leydig Cells/metabolism , Male , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Testis/cytology , Testis/metabolism , Testis/pathology , Transforming Growth Factor beta1/metabolism , bcl-2-Associated X Protein/metabolism
8.
Mol Cell Endocrinol ; 538: 111469, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34601003

ABSTRACT

The study aimed to analyze the time-dependent consequences of stress on gene expression responsible for diurnal endocrine Leydig cell function connecting them to the glucocorticoid-signaling. In the first 24h after the stress event, a daily variation of blood corticosterone increased, and testosterone decreased; the testosterone/corticosterone were lowest at the end of the stress session overlapping with inhibition of Leydig cells' steroidogenesis-related genes (Nr3c1/GR, Hsd3b1/2, Star, Cyp17a1) and changed circadian activity of the clock genes (the increased Bmal1/BMAL1 and Per1/2/PER1 and decreased Cry1 and Rev-erba). The glucocorticoid-treated rats showed a similar response. The principal-component-analysis (PCA) displayed an absence of significant differences between treatments especially on Per1 and Rev-erba, the findings confirmed by the in vivo blockade of the testicular glucocorticoid receptor (GR) during stress and ex vivo treatment of the Leydig cells with hydrocortisone and GR-blocker. In summary, stressful stimuli can entrain the clock in the Leydig cells through glucocorticoid-mediated communication.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/genetics , Corticosterone/blood , Leydig Cells/metabolism , Testosterone/blood , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Circadian Rhythm , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Leydig Cells/cytology , Male , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Stress, Physiological
9.
Cells ; 10(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34359871

ABSTRACT

The creation of a testis organoid (artificial testis tissue) with sufficient resemblance to the complex form and function of the innate testis remains challenging, especially using non-rodent donor cells. Here, we report the generation of an organoid culture system with striking biomimicry of the native immature testis tissue, including vasculature. Using piglet testis cells as starting material, we optimized conditions for the formation of cell spheroids, followed by long-term culture in an air-liquid interface system. Both fresh and frozen-thawed cells were fully capable of self-reassembly into stable testis organoids consisting of tubular and interstitial compartments, with all major cell types and structural details expected in normal testis tissue. Surprisingly, our organoids also developed vascular structures; a phenomenon that has not been reported in any other culture system. In addition, germ cells do not decline over time, and Leydig cells release testosterone, hence providing a robust, tunable system for diverse basic and applied applications.


Subject(s)
Biomimetic Materials/pharmacology , Organoids/physiology , Testis/blood supply , Animals , Cell Count , Cryopreservation , Leydig Cells/cytology , Leydig Cells/drug effects , Luteinizing Hormone/metabolism , Male , Neovascularization, Physiologic/drug effects , Organ Specificity , Organoids/cytology , Organoids/drug effects , Organoids/ultrastructure , Swine , Testis/cytology , Testis/ultrastructure , Testosterone/metabolism
10.
Gene ; 804: 145897, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34418471

ABSTRACT

This study was to explore the regulation effect of PGAM1 on the proliferation, apoptosis and glycolysis pathway of Tibetan sheep Sertoli cells. In this paper, the reproductive organs of male Tibetan sheep before pre-puberty (3 months old), sexual maturity (1 year old) and adult (3 years old) were used as experimental materials. The complete CDS region sequence of PGAM1 gene was cloned for bioinformatics analysis, and had the closest relationship with Tibetan antelope. QRT-PCR, Western blot and immunohistochemical staining were used to detect the expression and localization of PGAM1 in the testis and epididymis tissues of Tibetan sheep at different growth and development stages at the transcription and translation levels. Then the Tibetan sheep primary Sertoli cells (SCs) were isolated to construct PGAM1 gene overexpression and interference vectors, and to transfect primary SCs so as to promote and inhibit PGAM1 gene expression; CCK-8 and flow cytometry were used to detect the proliferation effect of SCs;qRT-PCR technology was employed to detect the changes in the expression of genes related to cell proliferation and apoptosis. Different kits were used to detect pyruvate, lactic acid, ATP production and LDH activity during glycolysis, and to detect the changes in the expression of downstream genes in the glycolysis pathway. The results showed that the CDS region of Tibetan sheep PGAM1 gene was 765 bp in length, which can encode 254 amino acids; and the expression of PGAM1 protein in the testis and epididymis increased at 1Y group and 3Ygroup compared with 3 M group, and that the PGAM1 protein mainly existed in SCs and Leydig cells at different developmental stages. CCK-8 and flow cytometry test results found that compared with the empty vector group (pcDNA3.1(+)), the proliferation rate of the PGAM1 gene overexpression group (pcDNA3.1(+)-PGAM1) decreased. The mRNA expression of the cell proliferation related genes PCNA and Bcl2 was significantly decreased (P < 0.05), and the expression of apoptosis-related genes Bax and caspase3 was significantly increased (P < 0.05). The expression of downstream genes in the glycolysis pathway was significant increased (P < 0.05), pyruvate content, ATP content, lactic acid production and LDH activity increased significantly (P < 0.05). Compared with the interference control group (NC), the proliferation rate of the PGAM1 gene interference group (si-PGAM1) was weakened. The mRNA expression of the cell proliferation-related genes PCNA and Bcl2 was significantly increased (P < 0.05), and the expression of cell apoptosis related genes Bax and caspase3 was significantly decreased (P < 0.05). The expression of downstream genes in the glycolysis pathway was significantly reduced (P < 0.05), and the pyruvate content, ATP content, lactic acid production and LDH activity were significantly decreased (P < 0.05). The PGAM1 gene might regulate the glycolytic metabolism pathway and regulate the sperm formation and maturation process by affecting the proliferation and apoptosis of SCs. This result provides basic data for the study of the function of PGAM1 in sheep testicular development.


Subject(s)
Phosphoglycerate Mutase/genetics , Phosphoglycerate Mutase/metabolism , Sertoli Cells/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Glycolysis/physiology , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Sertoli Cells/cytology , Sex Differentiation/genetics , Sexual Maturation/genetics , Sexual Maturation/physiology , Sheep/metabolism , Testis/metabolism
11.
J Steroid Biochem Mol Biol ; 214: 105974, 2021 11.
Article in English | MEDLINE | ID: mdl-34425195

ABSTRACT

Sex steroids play critical roles in sex differentiation and gonadal development in teleosts. Steroidogenic acute regulatory protein (StAR), transporting cholesterol (the substrate for steroidogenesis) from the outer mitochondrial membrane to the inner membrane, is the first rate-limiting factor of steroidogenesis. Interestingly, two StAR genes (named as StAR1 and StAR2) have been isolated from non-mammalian vertebrates. To characterize the functions of the novel StAR2 gene in the gonadal differentiation and fertility, we generated a StAR2 homozygous mutant line in Nile tilapia (Oreochromis niloticus). StAR2 gene knockout in male tilapia impeded meiotic initiation, associate with the down-regulation of meiosis related gene expressions of vasa, sycp3 and dazl at 90 days after hatching (dah). Meanwhile, cyp11b2 expression and serum 11-KT production significantly declined in StAR2-/- XY fish at 90 dah. From 120-300 dah, spermatogenesis gradually recovered, and so did the expressions of vasa, sycp3 and dazl in StAR2-/- XY fish testes. However, seminiferous lobules arranged disorderly in StAR2-/- XY fish testes at 300 dah. The number of Leydig cells and expressions of downstream steroidogenesis enzymes including cyp11a1, 3ß-HSD-I, 3ß-HSD-II, cyp17a1 and cyp17a2 decreased in StAR2-/- XY fish testes at 300 dah. Serum testosterone and 11-KT levels were significantly lower in StAR2-/- XY fish than that of their control counterparts. Furthermore, significantly elevated ar, fsh and lh expressions in StAR2-deficient XY fish testes and pituitaries were found when compared with the control XY fish. Testes degeneration and spermatogenic cell apoptosis were observed, while no sperm were squeezed out in StAR2-/- XY fish testes at 540 dah. Taken together, our results suggest that StAR2 has a role in testicular development, spermatogenesis and spermiation by regulating androgen production in tilapia, but may not be essential and could be compensated.


Subject(s)
Cell Differentiation , Cichlids/metabolism , Phosphoproteins/physiology , RNA-Binding Proteins/physiology , Spermatogenesis , Testis/embryology , Androgens/metabolism , Animals , Apoptosis , CRISPR-Cas Systems , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gonads/metabolism , Leydig Cells/cytology , Male , Mutation , Spermatozoa/physiology , Testis/metabolism , Testosterone/metabolism
12.
Mol Cell Endocrinol ; 535: 111389, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34229003

ABSTRACT

Effective procedures for the purification of Leydig cells (LCs) can facilitate functional studies and transplantation therapies. However, current methods to purify LCs from testes are still far from satisfactory. Here, we found that testicular autofluorescence existed in the interstitium along with the gradual maturation of LCs from birth to adulthood. These autofluorescent cells were further isolated by fluorescence-activated cell sorting (FACS) and determined to be composed of LCs and macrophages. To further purify LCs, we combined two fluorescence channels of FACS and successfully separated LCs and macrophages. Of note, we confirmed that the obtained LCs not only possessed high purity, viability and quantity but also had intact steroidogenic activity and excellent responsiveness to luteinizing hormone. Moreover, subcutaneous transplantation of isolated LCs could alleviate the symptoms of testosterone deficiency in castrated mice. In summary, we established an effective autofluorescence-based method for isolating LCs. This method will aid in the future success of using LCs for basic and translational applications.


Subject(s)
Cell Separation/methods , Leydig Cells/cytology , Leydig Cells/transplantation , Orchiectomy/adverse effects , Testosterone/metabolism , Animals , Cell Survival , Cells, Cultured , Disease Models, Animal , Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Optical Imaging , Testosterone/deficiency
13.
Cell Tissue Res ; 386(2): 379-389, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34278519

ABSTRACT

This study aimed to investigate the role of lncRNA FENDRR in apoptosis of Leydig cells and the further mechanism. The apoptosis of Leydig cells (TM3 cell line) was induced by H2O2-treatment and detected by flow cytometry. The function of FENDRR was determined by in vitro and in vivo silencing experiments. The mechanism of FENDRR in regulating the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) was assessed by RNA immunoprecipitation, RNA pull-down, and ubiquitination assays. FENDRR expression was up-regulated in H2O2-treated TM3 cells. Knockdown of FENDRR augmented Nrf2 and HO-1 protein levels and testosterone production in H2O2-treated TM3 cells, whereas the apoptosis rate and caspase 3 activity were decreased. Mechanically, FENDRR bound to Nrf2 and promoted its ubiquitination and degradation. Nrf2 overexpression reversed the effects FENDRR overexpression on apoptosis, caspase 3 activity, and testosterone concentration in H2O2-treated TM3 cells. The in vivo experiments showed that FENDRR silence increased serum testosterone level and improved testosterone-related anti-depression behaviors of late-onset hypogonadism (LOH) mice. Our findings suggested that FENDRR could promote apoptosis of Leydig cells in LOH partly through facilitating Nrf2 degradation.


Subject(s)
Hypogonadism/genetics , Leydig Cells/cytology , NF-E2-Related Factor 2/genetics , RNA, Long Noncoding/genetics , Animals , Apoptosis , Cell Line , Gene Knockdown Techniques , Hypogonadism/metabolism , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Proteolysis , Up-Regulation
14.
Nat Commun ; 12(1): 3876, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162856

ABSTRACT

Testicular development and function rely on interactions between somatic cells and the germline, but similar to other organs, regenerative capacity declines in aging and disease. Whether the adult testis maintains a reserve progenitor population remains uncertain. Here, we characterize a recently identified mouse testis interstitial population expressing the transcription factor Tcf21. We found that TCF21lin cells are bipotential somatic progenitors present in fetal testis and ovary, maintain adult testis homeostasis during aging, and act as potential reserve somatic progenitors following injury. In vitro, TCF21lin cells are multipotent mesenchymal progenitors which form multiple somatic lineages including Leydig and myoid cells. Additionally, TCF21+ cells resemble resident fibroblast populations reported in other organs having roles in tissue homeostasis, fibrosis, and regeneration. Our findings reveal that the testis, like other organs, maintains multipotent mesenchymal progenitors that can be potentially leveraged in development of future therapies for hypoandrogenism and/or infertility.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Homeostasis/genetics , Mesenchymal Stem Cells/metabolism , Regeneration/genetics , Testis/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage/genetics , Cells, Cultured , Female , Gene Expression Profiling/methods , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Mice, Transgenic , Single-Cell Analysis/methods , Testis/cytology
15.
Nat Rev Urol ; 18(8): 487-507, 2021 08.
Article in English | MEDLINE | ID: mdl-34188209

ABSTRACT

In Leydig cell dysfunction, cells respond weakly to stimulation by pituitary luteinizing hormone, and, therefore, produce less testosterone, leading to primary hypogonadism. The most widely used treatment for primary hypogonadism is testosterone replacement therapy (TRT). However, TRT causes infertility and has been associated with other adverse effects, such as causing erythrocytosis and gynaecomastia, worsening obstructive sleep apnoea and increasing cardiovascular morbidity and mortality risks. Stem-cell-based therapy that re-establishes testosterone-producing cell lineages in the body has, therefore, become a promising prospect for treating primary hypogonadism. Over the past two decades, substantial advances have been made in the identification of Leydig cell sources for use in transplantation surgery, including the artificial induction of Leydig-like cells from different types of stem cells, for example, stem Leydig cells, mesenchymal stem cells, and pluripotent stem cells (PSCs). PSC-derived Leydig-like cells have already provided a powerful in vitro model to study the molecular mechanisms underlying Leydig cell differentiation and could be used to treat men with primary hypogonadism in a more specific and personalized approach.


Subject(s)
Androgens/therapeutic use , Hypogonadism/therapy , Hypothalamo-Hypophyseal System/metabolism , Leydig Cells/metabolism , Reproductive Control Agents/therapeutic use , Stem Cell Transplantation , Testis/metabolism , Adult Stem Cells , Animals , Chorionic Gonadotropin/therapeutic use , Embryonic Stem Cells , Hormone Replacement Therapy , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells , Leydig Cells/cytology , Leydig Cells/transplantation , Luteinizing Hormone/therapeutic use , Male , Mesenchymal Stem Cells , Testis/cytology , Testosterone/therapeutic use
16.
Front Endocrinol (Lausanne) ; 12: 599149, 2021.
Article in English | MEDLINE | ID: mdl-33815270

ABSTRACT

The function of immature Leydig cells is regulated by hormones, such as androgen and luteinizing hormone (LH). However, the regulation of this process is still unclear. The objective of this study was to determine whether luteinizing hormone (LH) or androgens contribute to this process. Immature Leydig cells were purified from 35-day-old male Sprague Dawley rats and cultured with LH (1 ng/ml) or androgen (7α-methyl-19- nortestosterone, MENT, 100 nM) for 2 days. LH or MENT treatment significantly increased the androgens produced by immature Leydig cells in rats. Microarray and qPCR and enzymatic tests showed that LH up-regulated the expression of Scarb1, Cyp11a1, Cyp17a1, and Srd5a1 while down-regulated the expression of Sult2a1 and Akr1c14. On the contrary, the expression of Cyp17a1 was up-regulated by MENT. LH and MENT regulate Leydig cell function through different sets of transcription factors. We conclude that LH and androgens participate in the regulation of rat immature Leydig cell function through different transcriptional pathways.


Subject(s)
Androgens/metabolism , Leydig Cells/metabolism , Luteinizing Hormone/metabolism , Nandrolone/analogs & derivatives , Animals , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Humans , Leydig Cells/cytology , Male , Nandrolone/metabolism , Rats , Rats, Sprague-Dawley , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Transcription, Genetic
17.
Food Chem Toxicol ; 151: 112152, 2021 May.
Article in English | MEDLINE | ID: mdl-33774092

ABSTRACT

Perfluorododecanoic acid (PFDoA) is an endocrine-damaging compound in contaminated food and water. However, the potential role and underlying mechanism of PFDoA in Leydig cell regeneration from stem Leydig cells remain unclear. The current study aims to investigate the effect of PFDoA on the regeneration of Leydig cells in the testis of rats treated with ethylene dimethane sulfonate (EDS). PFDoA (0, 5 or 10 mg/kg/day) was gavaged to adult Sprague-Dawley male rats for 8 days, and 75 mg/kg EDS was intraperitoneally injected to eliminate Leydig cells to initiate its regeneration from day 21-56 after EDS. The serum testosterone levels in the 5 and 10 mg/kg/day PFDoA groups were significantly reduced at day 21 after EDS and the levels of serum luteinizing hormone and follicle-stimulating hormone were significantly decreased in the 10 mg/kg/day PFDoA groups at day 56 after EDS. PFDoA significantly reduced Leydig cell number and proliferation at a dose of 10 mg/kg at days 21 and 56 after EDS. PFDoA significantly down-regulated the expression of Leydig cell-specific genes (Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1 and Cyp17a1) and their proteins at both doses at days 21 and 56 after EDS. PFDoA significantly down-regulated the gene expression of Sertoli cells (Fshr, Dhh, and Sox9) at 5 mg/kg or higher at days 21 and 56 after EDS. In addition, we found that PFDoA significantly inhibited EdU incorporation into putative stem Leydig cells and their differentiation into the Leydig cell lineage in vitro. In conclusion, short-term PFDoA exposure in adulthood delayed the regeneration of Leydig cells by preventing Leydig cells from stem cells via multiple mechanisms.


Subject(s)
Cell Proliferation/drug effects , Lauric Acids/toxicity , Leydig Cells/drug effects , Stem Cells/drug effects , Animals , Body Weight/drug effects , Cell Differentiation/drug effects , Fluorocarbons , Histones/metabolism , Humans , Leydig Cells/cytology , Male , Methylation , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Sperm Count , Stem Cells/cytology , Testis/drug effects , Testosterone/blood , Testosterone/metabolism
18.
Reproduction ; 161(5): 523-537, 2021 05.
Article in English | MEDLINE | ID: mdl-33730690

ABSTRACT

Long ncRNAs regulate a complex array of fundamental biological processes, while its molecular regulatory mechanism in Leydig cells (LCs) remains unclear. In the present study, we established the lncRNA LOC102176306/miR-1197-3p/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PPARGC1A) regulatory network by bioinformatic prediction, and investigated its roles in goat LCs. We found that lncRNA LOC102176306 could efficiently bind to miR-1197-3p and regulate PPARGC1A expression in goat LCs. Downregulation of lncRNA LOC102176306 significantly supressed testosterone (T) synthesis and ATP production, decreased the activities of antioxidant enzymes and mitochondrial complex I and complex III, caused the loss of mitochondrial membrane potential, and inhibited the proliferation of goat LCs by decreasing PPARGC1A expression, while these effects could be restored by miR-1197-3p inhibitor treatment. In addition, miR-1197-3p mimics treatment significantly alleviated the positive effects of lncRNA LOC102176306 overexpression on T and ATP production, antioxidant capacity and proliferation of goat LCs. Taken together, lncRNA LOC102176306 functioned as a sponge for miR-1197-3p to maintain PPARGC1A expression, thereby affecting the steroidogenesis, cell proliferation and oxidative stress of goat LCs. These findings extend our understanding of the molecular mechanisms of T synthesis, cell proliferation and oxidative stress of LCs.


Subject(s)
Leydig Cells/cytology , MicroRNAs/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Long Noncoding/genetics , Testis/cytology , Animals , Apoptosis , Cell Proliferation , Goats , Leydig Cells/metabolism , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Testis/metabolism , Testosterone/metabolism
19.
Endocrinology ; 162(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33507237

ABSTRACT

Type 1 diabetes mellitus (T1DM) is associated with impaired spermatogenesis and lower testosterone levels and epididymal weight. However, the underlying processes in the testis are unknown and remain to be elucidated. Therefore, the present study focused on the effects of T1DM on testicular function in a spontaneously diabetic rat model. BB/OKL rats after diabetes manifestation were divided into 3 groups: those without insulin treatment and insulin treatment for a duration of 2 and of 6 weeks. Anthropometrical data, circulating levels of gonadotrophins, testosterone, and inhibin B were measured. Intratesticular testosterone, oxidative stress, inflammation, and apoptosis were analyzed. Key enzymes of steroidogenesis were evaluated in the testis. Untreated diabetic rats had significantly lower serum follicle-stimulating hormone and luteinizing hormone levels. Serum and intratesticular testosterone levels significantly decreased in untreated diabetic rats compared to healthy controls. Key markers of Leydig cell function were significantly downregulated at the RNA level: insulin-like factor 3 (Insl3) by 53% (P = .006), Star by 51% (P = .004), Cyp11A1 by 80% (P = .003), 3Beta-Hsd2 by 61% (P = .005), and Pbr by 52% (P = .002). In the insulin-treated group, only Cyp11A1 and 3Beta-Hsd2 transcripts were significantly lower. Interestingly, the long-term insulin-treated group showed significant upregulation of most steroidogenic enzymes without affecting testosterone levels. Tumor necrosis factor α and apoptosis were significantly increased in the long-term insulin-treated rats. In conclusion T1DM, with a severe lack of insulin, has an adverse action on Leydig cell function. This is partially reversible with well-compensated blood glucose control. Long-term T1DM adversely affects Leydig cell function because of the process of inflammation and apoptosis.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Insulin/administration & dosage , Leydig Cells/drug effects , Leydig Cells/metabolism , Animals , Apoptosis/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Follicle Stimulating Hormone/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Leydig Cells/cytology , Luteinizing Hormone/metabolism , Male , Proteins/genetics , Proteins/metabolism , Rats , Spermatogenesis/drug effects , Testis/cytology , Testis/drug effects , Testis/metabolism , Testosterone/metabolism
20.
Mol Cell Endocrinol ; 525: 111179, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33515640

ABSTRACT

Peritubular stem Leydig cells (SLCs) have been identified from rat testicular seminiferous tubules. However, no stem cells for peritubular myoid cells have been reported in the adult testis so far. In the present study, we tested the hypothesis that the peritubular SLCs are multipotent and able to form either Leydig or myoid cells. Using cultured tubules, we show that in the presence of PDGFAA and luteinizing hormone, SLCs became testosterone-producing Leydig cells, while in the presence of PDGFBB and TGFB, the cells formed α-smooth muscle actin-expressing myoid cells. This multipotency was also confirmed by culture of isolated CD90+ SLCs. These results suggest that these stem cells outside the myoid layer are multipotent and give rise to either Leydig or myoid cells, depending on the inducing factors. These cells may serve as a common precursor population for maintaining homeostasis of both Leydig and myoid cell populations in the adult testis.


Subject(s)
Cell Differentiation , Cell Lineage , Leydig Cells/cytology , Seminiferous Tubules/cytology , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Leydig Cells/drug effects , Male , Platelet-Derived Growth Factor/pharmacology , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/metabolism , Thy-1 Antigens/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...