Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell Mol Life Sci ; 81(1): 93, 2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38367191

RESUMO

Stem Leydig cells (SLCs) are essential for maintaining normal spermatogenesis as the significant component of testis microenvironment and gonadal aging. Although progress has been achieved in the regulation of male germ cells in mammals and humans, it remains unknown about the genes and signaling pathways of human SLCs. Here we have demonstrated, for the first time, that WNT5A (Wnt family member 5a) mediates the proliferation, apoptosis, and stemness of human SLCs, namely NGFR+ Leydig cells. We revealed that NGFR+ Leydig cells expressed NGFR, PDGFRA, NES, NR2F2, and THY1, hallmarks for SLCs. RNA-sequencing showed that WNT5A was expressed at a higher level in human SLCs than non-SLCs, while immunohistochemistry and Western blots further illustrated that WNT5A was predominantly expressed in human SLCs. Notably, CCK-8, EdU and Western blots displayed that WNT5A enhanced the proliferation and DNA synthesis and retained stemness of human SLCs, whereas flow cytometry and TUNEL analyses demonstrated that WNT5A inhibited the apoptosis of these cells. WNT5A knockdown caused an increase in LC lineage differentiation of human SLCs and reversed the effect of WNT5A overexpression on fate decisions of human SLCs. In addition, WNT5A silencing  resulted in the decreases in nuclear translocation of ß-catenin and expression levels of c-Myc, CD44, and Cyclin D1. Collectively, these results implicate that WNT5A regulates the proliferation, apoptosis and stemness of human SLCs through the activation of the ß-catenin signaling pathway. This study thus provides a novel molecular mechanism underlying the fate determinations of human SLCs, and it offers a new insight into the niche regulation of human testis.


Assuntos
Células Intersticiais do Testículo , beta Catenina , Animais , Humanos , Masculino , Células Intersticiais do Testículo/metabolismo , beta Catenina/metabolismo , Testículo/metabolismo , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Transdução de Sinais , Apoptose , Proliferação de Células , Via de Sinalização Wnt/genética , Mamíferos/metabolismo
2.
Semin Cell Dev Biol ; 121: 63-70, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34001436

RESUMO

Stem cells are specialized cells that can renew themselves through cell division and can differentiate into multi-lineage cells. Mesenchymal stem cells are adult stem cells that exist in animal and human tissues. Mesenchymal stem cells have the ability to differentiate into mesodermal lineages, such as Leydig cells, adipocytes, osteocytes, and chondrocytes. Mesenchymal stem cells express cell surface markers, such as cluster of differentiation (CD) 29, CD44, CD73, CD90, CD105, and lack the expression of CD14, CD34, CD45 and HLA (human leukocyte antigen)-DR. Stem Leydig cells are one kind of mesenchymal stem cells, which are present in the interstitial compartment of testis. Stem Leydig cells are multipotent and can differentiate into Leydig cells, adipocytes, osteocytes, and chondrocytes. Stem Leydig cells have been isolated from rodent and human testes. Stem Leydig cells may have potential therapeutic values in several clinical applications, such as the treatment of male hypogonadism and infertility. In this review, we focus on the latest research on stem Leydig cells of both rodents and human, the expression of cell surface markers, culture, differentiation potential, and their applications.


Assuntos
Células Intersticiais do Testículo/metabolismo , Medicina Regenerativa/métodos , Saúde Reprodutiva/normas , Animais , Humanos , Masculino , Camundongos , Ratos
3.
BMC Dev Biol ; 20(1): 20, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33023470

RESUMO

BACKGROUND: Monocyte chemoattractant protein-1(MCP-1) is a chemokine secreted by Leydig cells and peritubular myoid cells in the rat testis. Its role in regulating the development of Leydig cells via autocrine and paracrine is still unclear. The objective of the current study was to investigate the effects of MCP-1 on Leydig cell regeneration from stem cells in vivo and on Leydig cell development in vitro. RESULTS: Intratesticular injection of MCP-1(10 ng/testis) into Leydig cell-depleted rat testis from post-EDS day 14 to 28 significantly increased serum testosterone and luteinizing hormone levels, up-regulated the expression of Leydig cell proteins, LHCGR, SCARB1, CYP11A1, HSD3B1, CYP17A1, and HSD17B3 without affecting progenitor Leydig cell proliferation, as well as increased ERK1/2 phosphorylation. MCP-1 (100 ng/ml) significantly increased medium testosterone levels and up-regulated LHCGR, CYP11A1, and HSD3B1 expression without affecting EdU incorporation into stem cells after in vitro culture for 7 days. RS102895, a CCR2 inhibitor, reversed MCP-1-mediated increase of testosterone level after culture in combination with MCP-1. CONCLUSION: MCP-1 stimulates the differentiation of stem and progenitor Leydig cells without affecting their proliferation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Quimiocina CCL2/farmacologia , Células Intersticiais do Testículo/citologia , Regeneração/efeitos dos fármacos , Células-Tronco/citologia , Testículo/fisiologia , Animais , Expressão Gênica/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/sangue , Masculino , Ratos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Testículo/efeitos dos fármacos , Testosterona/sangue
4.
Hum Reprod ; 35(10): 2197-2212, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32951040

RESUMO

STUDY QUESTION: Is endosialin a specific marker of human stem Leydig cells (SLCs) with the ability to differentiate into testosterone-producing Leydig cells (LCs) in vitro and in vivo? SUMMARY ANSWER: Endosialin is a specific marker of human SLCs which differentiate into testosterone-producing LCs in vitro and in vivo. WHAT IS KNOWN ALREADY: Human SLCs have been identified and isolated using the marker platelet-derived growth factor receptor α (PDGFRα) or nerve growth factor receptor (NGFR). However, the specificity was not high; thus, LCs and germ cells could be mistakenly sorted as SLCs if PDGFRα or NGFR was used as a marker for human SLCs isolation. STUDY DESIGN, SIZE, DURATION: Firstly, we re-evaluated the specificity of PDGFRα and NGFR for SLCs in adult human testes. Then we analysed the previously published single-cell sequencing data and found that endosialin may identify human SLCs. Subsequently, we sorted endosialin+ cells from four human donors and characterized their self-renewal and multipotent properties. To assess whether endosialin+ cells have the potential to differentiate into functional LCs in vitro, these cells were stimulated by differentiation-inducing medium. We next assessed the in vivo regenerative potential of human endosialin+ cells after xenotransplantation into the testes of immunodeficient mice. PARTICIPANTS/MATERIALS, SETTING, METHODS: Single-cell sequencing analysis, immunofluorescence and flow cytometry were used to characterize human testis tissues. In vitro colony formation, multipotent differentiation (adipogenic, osteogenic and chondrogenic) and Leydig cell-lineage induction were used to assess stem cell activity. Xenotransplantation into 3-week-old immunodeficient mice was used to determine in vivo regenerative potential. Endpoint measures included testosterone measurements, cell proliferation, immunofluorescence, flow cytometry and quantitative RT-PCR. MAIN RESULTS AND THE ROLE OF CHANCE: The results indicate that endosialin is a specific marker of SLCs compared with PDGFRα and NGFR. Additionally, endosialin+ cells isolated from human testes show extensive proliferation and differentiation potential in vitro: their self-renewal ability was inferred by the formation of spherical clones derived from a single cell. Moreover, these cells could differentiate into functional LCs that secreted testosterone in response to LH in a concentration-dependent manner in vitro. These self-renewal and differentiation properties reinforce the proposal that human testicular endosialin+ cells are SLCs. Furthermore, transplanted human endosialin+ cells appear to colonize the murine host testes, localize to peritubular and perivascular regions, proliferate measurably and differentiate partially into testosterone-producing LCs in vivo. LARGE SCALE DATA: NA. LIMITATIONS, REASONS FOR CAUTION: Owing to the difficulty in collecting human testis tissue, the sample size was limited. The functions of endosialin on SLCs need to be elucidated in future studies. WIDER IMPLICATIONS OF THE FINDINGS: A discriminatory marker, endosialin, for human SLCs purification is a prerequisite to advance research in SLCs and logically promote further clinical translation of SLCs-based therapies for male hypogonadism. STUDY FUNDING/COMPETING INTEREST(S): A.P.X. was supported by the National Key Research and Development Program of China (2017YFA0103802 and 2018YFA0107200). C.D. was supported by the National Natural Science Foundation of China (81971314) and the Natural Science Foundation of Guangdong Province, China (2018B030311039). The authors declare no conflict of interest.


Assuntos
Células Intersticiais do Testículo , Testículo , Adulto , Animais , Diferenciação Celular , China , Humanos , Masculino , Camundongos , Células-Tronco , Testosterona
5.
J Cell Mol Med ; 23(1): 426-438, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30320465

RESUMO

Oncostatin M (OSM) is a pleiotropic cytokine within the interleukin six family of cytokines, which regulate cell growth and differentiation in a wide variety of biological systems. However, its action and underlying mechanisms on stem Leydig cell development are unclear. The objective of the present study was to investigate whether OSM affects the proliferation and differentiation of rat stem Leydig cells. We used a Leydig cell regeneration model in rat testis and a unique seminiferous tubule culture system after ethane dimethane sulfonate (EDS) treatment to assess the ability of OSM in the regulation of proliferation and differentiation of rat stem Leydig cells. Intratesticular injection of OSM (10 and 100 ng/testis) from post-EDS day 14 to 28 blocked the regeneration of Leydig cells by reducing serum testosterone levels without affecting serum luteinizing hormone and follicle-stimulating hormone levels. It also decreased the levels of Leydig cell-specific mRNAs (Lhcgr, Star, Cyp11a1, Hsd3b1, Cyp17a1 and Hsd11b1) and their proteins by the RNA-Seq and Western blotting analysis. OSM had no effect on the proliferative capacity of Leydig cells in vivo. In the seminiferous tubule culture system, OSM (0.1, 1, 10 and 100 ng/mL) inhibited the differentiation of stem Leydig cells by reducing medium testosterone levels and downregulating the expression of Leydig cell-specific genes (Lhcgr, Star, Cyp11a1, Hsd3b1, Cyp17a1 and Hsd11b1) and their proteins. OSM-mediated action was reversed by S3I-201 (a STAT3 antagonist) or filgotinib (a JAK1 inhibitor). These data suggest that OSM is an inhibitory factor of rat stem Leydig cell development.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos dos fármacos , Oncostatina M/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Hormônio Foliculoestimulante/metabolismo , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testosterona/metabolismo
6.
Stem Cells ; 35(5): 1222-1232, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28090714

RESUMO

Stem Leydig cell (SLC) transplantation could provide a new strategy for treating the testosterone deficiency. Our previous study demonstrated that CD51 (also called integrin αv) might be a putative cell surface marker for SLCs, but the physiological function and efficacy of CD51+ SLCs treatment remain unclear. Here, we explore the potential therapeutic benefits of CD51+ SLCs transplantation and whether these transplanted cells can be regulated by the hypothalamic-pituitary-gonadal (HPG) axis. CD51+ cells were isolated from the testes of 12-weeks-old C57BL/6 mice, and we showed that such cells expressed SLC markers and that they were capable of self-renewal, extensive proliferation, and differentiation into multiple mesenchymal cell lineages and LCs in vitro. As a specific cytotoxin that eliminates Leydig cells (LCs) in adult rats, ethane dimethanesulfonate (EDS) was used to ablate LCs before the SLC transplantation. After being transplanted into the testes of EDS-treated rats, the CD51+ cells differentiated into mature LCs, and the recipient rats showed a partial recovery of testosterone production and spermatogenesis. Notably, a testosterone analysis revealed a circadian rhythm of testosterone secretion in cell-transplanted rats, and these testosterone secretions could be suppressed by decapeptyl (a luteinizing hormone-releasing hormone agonist), suggesting that the transplanted cells might be regulated by the HPG axis. This study is the first to demonstrate that CD51+ SLCs can restore the neuroendocrine regulation of testicular function by physiologically recovering the expected episodic changes in diurnal testosterone serum levels and that SLC transplantation may provide a new tool for the studies of testosterone deficiency treatment. Stem Cells 2017;35:1222-1232.


Assuntos
Integrina alfaV/metabolismo , Células Intersticiais do Testículo/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Testosterona/deficiência , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Separação Celular , Modelos Animais de Doenças , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Mesilatos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Ratos Sprague-Dawley , Espermatogênese , Testículo/citologia
7.
Adv Sci (Weinh) ; 10(29): e2300993, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37697644

RESUMO

Hereditary primary hypogonadism (HPH), caused by gene mutation related to testosterone synthesis in Leydig cells, usually impairs male sexual development and spermatogenesis. Genetically corrected stem Leydig cells (SLCs) transplantation may provide a new approach for treating HPH. Here, a novel nonsense-point-mutation mouse model (LhcgrW495X ) is first generated based on a gene mutation relative to HPH patients. To verify the efficacy and feasibility of SLCs transplantation in treating HPH, wild-type SLCs are transplanted into LhcgrW495X mice, in which SLCs obviously rescue HPH phenotypes. Through comparing several editing strategies, optimized PE2 protein (PEmax) system is identified as an efficient and precise approach to correct the pathogenic point mutation in Lhcgr. Furthermore, delivering intein-split PEmax system via lentivirus successfully corrects the mutation in SLCs from LhcgrW495X mice ex vivo. Gene-corrected SLCs from LhcgrW495X mice exert ability to differentiate into functional Leydig cells in vitro. Notably, the transplantation of gene-corrected SLCs effectively regenerates Leydig cells, recovers testosterone production, restarts sexual development, rescues spermatogenesis, and produces fertile offspring in LhcgrW495X mice. Altogether, these results suggest that PE-based gene editing in SLCs ex vivo is a promising strategy for HPH therapy and is potentially leveraged to address more hereditary diseases in reproductive system.


Assuntos
Hipogonadismo , Células Intersticiais do Testículo , Receptores do LH , Animais , Humanos , Masculino , Camundongos , Diferenciação Celular , Hipogonadismo/genética , Hipogonadismo/terapia , Células Intersticiais do Testículo/transplante , Mutação , Receptores Acoplados a Proteínas G , Testosterona/metabolismo , Receptores do LH/genética
8.
Andrology ; 11(7): 1495-1513, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37029531

RESUMO

BACKGROUND: Ciliary neurotrophic factor is a member of the interleukin-6 family of cytokines. Ciliary neurotrophic factor drives many cells for their development. However, its effects on Leydig cell development remain unclear. METHODS: In the current study, we used three-dimensional seminiferous tubule culture system to induce the proliferation and differentiation of tubule-associated stem Leydig cells and primary progenitor Leydig cells culture to address the effects of ciliary neurotrophic factor. RESULTS: We found that ciliary neurotrophic factor stimulated the proliferation of stem Leydig cells but inhibited their development into the Leydig cell lineage. The ciliary neurotrophic factor-mediated effects can be reversed by signal transducer and activator 3 inhibitor S3I-201 and phosphatidylinositol 3-kinase inhibitor wortmannin, indicating that ciliary neurotrophic factor acts via signal transducer and activator 3-phosphatidylinositol 3-kinase signaling pathways to increase stem/progenitor Leydig cell proliferation. Ciliary neurotrophic factor at 1 and 10 ng/mL significantly decreased androgen production by progenitor Leydig cells. Microarray analysis of ciliary neurotrophic factor-treated progenitor Leydig cells showed that ciliary neurotrophic factor blocked steroidogenic pathways by downregulating Scarb1, Star, and Hsd3b1, possibly by downregulating the transcription factor Nr5a1 expression. CONCLUSION: Ciliary neurotrophic factor stimulates proliferation but blocks the differentiation of stem/progenitor Leydig cells.


Assuntos
Fator Neurotrófico Ciliar , Células Intersticiais do Testículo , Masculino , Ratos , Animais , Fator Neurotrófico Ciliar/farmacologia , Fator Neurotrófico Ciliar/metabolismo , Diferenciação Celular , Células Intersticiais do Testículo/metabolismo , Regulação da Expressão Gênica , Proliferação de Células
9.
Front Endocrinol (Lausanne) ; 14: 1139281, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37051204

RESUMO

Background: Testosterone plays a critical role in maintaining reproductive functions and well-beings of the males. Adult testicular Leydig cells (LCs) produce testosterone and are generated from stem Leydig cells (SLCs) during puberty through adulthood. In addition, macrophages are critical in the SLC regulatory niche for normal testicular function. Age-related reduction in serum testosterone contributes to a number of metabolic and quality-of-life changes in males, as well as age-related changes in immunological functions. How aging and testicular macrophages may affect SLC function is still unclear. Methods: SLCs and macrophages were purified from adult and aged mice via FACS using CD51 as a marker protein. The sorted cells were first characterized and then co-cultured in vitro to examine how aging and macrophages may affect SLC proliferation and differentiation. To elucidate specific aging effects on both cell types, co-culture of sorted SLCs and macrophages were also carried out across two ages. Results: CD51+ (weakly positive) and CD51++ (strongly positive) cells expressed typical SLC and macrophage markers, respectively. However, with aging, both cell types increased expression of multiple cytokine genes, such as IL-1b, IL-6 and IL-8. Moreover, old CD51+ SLCs reduced their proliferation and differentiation, with a more significant reduction in differentiation (2X) than proliferation (30%). Age matched CD51++ macrophages inhibited CD51+ SLC development, with a more significant reduction in old cells (60%) than young (40%). Crossed-age co-culture experiments indicated that the age of CD51+ SLCs plays a more significant role in determining age-related inhibitory effects. In LC lineage formation, CD51+ SLC had both reduced LC lineage markers and increased myoid cell lineage markers, suggesting an age-related lineage shift for SLCs. Conclusion: The results suggest that aging affected both SLC function and their regulatory niche cell, macrophages.


Assuntos
Maturidade Sexual , Testosterona , Masculino , Camundongos , Animais , Testosterona/metabolismo , Diferenciação Celular , Envelhecimento , Proliferação de Células , Macrófagos/metabolismo
10.
J Steroid Biochem Mol Biol ; 233: 106344, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37286111

RESUMO

Platelet-derived growth factor BB (BB) regulates cell proliferation and function. However, the roles of BB on proliferation and function of Leydig stem (LSCs) and progenitor cells (LPCs) and the underlying signaling pathways remain unclear. This study aimed to analyze the roles of PI3K and MAPK pathways in the regulation of proliferation-related and steroidogenesis-related gene expression in rat LSCs/LPCs. In this experiment, BB receptor antagonist, tyrosine kinase inhibitor IV (PKI), the PI3K inhibitor, LY294002, and the MEK inhibitor, U0126, were used to measure the effects of these pathways on the expression of cell cycle-related genes (Ccnd1 and Cdkn1b) and steroidogenesis-related genes (Star, Cyp11a1, Hsd3b1, Cyp17a1, and Srd5a1), as well as Leydig cell maturation gene Pdgfra [1]. These results showed that BB (10 ng/mL)-stimulated EdU-incorporation into LSCs and BB-mediated inhibition on its differentiation was mediated through the activation of its receptor, PDGFRB, as well as MAPK and PI3K pathways. The results of LPC experiment also showed that LY294002 and U0126 decreased BB (10 ng/mL)-upregulated Ccnd1 expression while only U0126 reversed BB (10 ng/mL)-downregulated Cdkn1b expression. U0126 significantly reversed BB (10 ng/mL)-mediated downregulation of Cyp11a1, Hsd3b1, and Cyp17a1 expression. On the other hand, LY294002 reversed the expression of Cyp17a1 and Abca1. In conclusion, BB-mediated induction of proliferation and suppression of steroidogenesis of LSCs/LPCs are dependent on the activation of both MAPK and PI3K pathways, which show distinct regulation of gene expression.


Assuntos
Enzima de Clivagem da Cadeia Lateral do Colesterol , Fosfatidilinositol 3-Quinases , Ratos , Masculino , Animais , Becaplermina/metabolismo , Becaplermina/farmacologia , Ratos Sprague-Dawley , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células Intersticiais do Testículo/metabolismo , Proliferação de Células , Transdução de Sinais
11.
Mol Cell Endocrinol ; 519: 111039, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32980418

RESUMO

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.


Assuntos
Células Imobilizadas/transplante , Células Intersticiais do Testículo/transplante , Testosterona/deficiência , Adipócitos/patologia , Adolescente , Adulto , Idoso , Alginatos/química , Antígenos CD/metabolismo , Medula Óssea/patologia , Cápsulas , Castração , Diferenciação Celular , Humanos , Células Intersticiais do Testículo/ultraestrutura , Masculino , Pessoa de Meia-Idade , Atrofia Muscular/patologia , Polilisina/análogos & derivados , Polilisina/química , Testosterona/metabolismo , Adulto Jovem
12.
Stem Cell Rev Rep ; 17(5): 1521-1533, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33598893

RESUMO

Male hypogonadism is a clinical syndrome caused by testosterone deficiency. Hypogonadism can be caused by testicular disease (primary hypogonadism) or hypothalamic-pituitary dysfunction (secondary hypogonadism). The present strategy for treating hypogonadism is the administration of exogenous testosterone. But exogenous testosterone is reported to have negative side effects including adverse cardiovascular events and disruption of physiological spermatogenesis probably due to its inability to mimic the physiological circadian rhythm of testosterone secretion in vivo. In recent years, a growing number of articles demonstrated that stem Leydig cells (SLCs) can not only differentiate into functional Leydig cells (LCs) in vivo to replace chemically disrupted LCs, but also secrete testosterone in a physiological pattern. The proliferation and differentiation of SLCs are regulated by various factors. However, the mechanisms involved in regulating the development of SLCs remain to be summarized. Factors involved in the regulation of SLCs can be divided into environmental pollutants, growth factors, cytokine and hormones. Environmental pollutants such as Perfluorooctanoic acid (PFOA) and Triphenyltin (TPT) could suppress SLCs proliferation or differentiation. Growth factors including FGF1, FGF16, NGF and activin A are essential for the maintenance of SLCs self-renewal and differentiation. Interleukin 6 family could inhibit differentiation of SLCs. Among hormones, dexamethasone suppresses SLCs differentiation, while aldosterone suppresses their proliferation. The present review focuses on new progress about factors regulating SLC's proliferation and differentiation which will undoubtedly deepen our insights into SLCs and help make better clinical use of them. Different factors affect on the proliferation and differentiation of stem Leydig cells. Firstly, each rat was intraperitoneally injected EDS so as to deplete Leydig cells from the adult testis. Secondly, the CD51+ or CD90+ cells from the testis of rats are SLCs, and the p75+ cells from human adult testes are human SLCs. These SLCs in the testis start to proliferate and some of them differentiate into LCs. Thirdly, during the SLCs regeneration period, researchers could explore different function of those factors (pollutants, growth factors, cytokines and hormones) towards SLCs.


Assuntos
Células Intersticiais do Testículo , Animais , Diferenciação Celular , Autorrenovação Celular , Poluentes Ambientais , Fatores de Crescimento de Fibroblastos , Hipogonadismo , Masculino , Testosterona
13.
Andrology ; 8(5): 1265-1276, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32416031

RESUMO

BACKGROUND: Origin of human adult Leydig cells (ALCs) is not well understood. This might be partly due to limited data available on the identification and location of human precursor and stem Leydig cells (SLCs) which hampers the study on the development of ALCs. OBJECTIVES: The aim of the present study was to investigate whether described human (PDGFRα, NGFR) and rodent (NES, PDGFRα, THY1, NR2F2) SLC markers are expressed by a common cell population within human adult testicular interstitial cells in vivo and before and after in vitro propagation. MATERIALS AND METHODS: Immunohistochemical analyses were used to identify localization of human adult testicular interstitial cells expressing described SLC markers. Next, interstitial cells were isolated and cultured. The percentage of cells expressing one or more SLC markers was determined before and after culture using flow cytometry. RESULTS: NR2F2 and PDGFRα were present in peritubular, perivascular, and Leydig cells, while THY1 was expressed in peritubular and perivascular cells. Although NES and NGFR were expressed in endothelial cells, co-localization with PDGFRα was found for both in vitro, although for NGFR only after culture. All marker positive cells were able to undergo propagation in vitro. DISCUSSION: The partly overlap in localization and overlap in expression in human testicular cells indicate that PDGFRα, NR2F2, and THY1 are expressed within the same ALC developmental lineage from SLCs. Based on the in vitro results, this is also true for NES and after in vitro propagation for NGFR. CONCLUSION: Our results that earlier described SLC markers are expressed in overlapping human interstitial cell population opens up further research strategies aiming for a better insight in the Leydig cell lineage and will be helpful for development of strategies to cure ALC dysfunction.


Assuntos
Biomarcadores/análise , Células Intersticiais do Testículo/citologia , Células-Tronco/citologia , Testículo/citologia , Linhagem da Célula , Humanos , Masculino
14.
Endocr Rev ; 41(1)2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31673697

RESUMO

Androgen deficiency (hypogonadism) affects males of all ages. Testosterone replacement therapy (TRT) is effective in restoring serum testosterone and relieving symptoms. TRT, however, is reported to have possible adverse effects in part because administered testosterone is not produced in response to the hypothalamic-pituitary-gonadal (HPG) axis. Progress in stem cell biology offers potential alternatives for treating hypogonadism. Adult Leydig cells (ALCs) are generated by stem Leydig cells (SLCs) during puberty. SLCs persist in the adult testis. Considerable progress has been made in the identification, isolation, expansion and differentiation of SLCs in vitro. In addition to forming ALCs, SLCs are multipotent, with the ability to give rise to all 3 major cell lineages of typical mesenchymal stem cells, including osteoblasts, adipocytes, and chondrocytes. Several regulatory factors, including Desert hedgehog and platelet-derived growth factor, have been reported to play key roles in the proliferation and differentiation of SLCs into the Leydig lineage. In addition, stem cells from several nonsteroidogenic sources, including embryonic stem cells, induced pluripotent stem cells, mature fibroblasts, and mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord have been transdifferentiated into Leydig-like cells under a variety of induction protocols. ALCs generated from SLCs in vitro, as well as Leydig-like cells, have been successfully transplanted into ALC-depleted animals, restoring serum testosterone levels under HPG control. However, important questions remain, including: How long will the transplanted cells continue to function? Which induction protocol is safest and most effective? For translational purposes, more work is needed with primate cells, especially human.


Assuntos
Células Intersticiais do Testículo/citologia , Células-Tronco/citologia , Testículo/citologia , Adulto , Animais , Diferenciação Celular , Linhagem da Célula/fisiologia , Humanos , Hipogonadismo/etiologia , Hipogonadismo/patologia , Hipogonadismo/terapia , Células Intersticiais do Testículo/fisiologia , Masculino , Espermatogênese/fisiologia , Células-Tronco/fisiologia , Testículo/fisiologia
15.
Mol Cell Endocrinol ; 481: 35-43, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30476560

RESUMO

We reported previously that stem Leydig cells (SLC) on the surfaces of rat testicular seminiferous tubules are able to differentiate into Leydig cells. The proliferation and differentiation of SLCs seem likely to be regulated by niche cells, including nearby germ and Sertoli cells. Due to the cyclical nature of spermatogenesis, we hypothesized that the changes in the germ cell composition of the seminiferous tubules as spermatogenesis proceeds may affect tubule-associated SLC functions. To test this hypothesis, we compared the ability of SLCs associated with tubules at different stages of the cycle to differentiate into Leydig cells in vitro. SLCs associated with stages IX-XI were more active in proliferation and differentiation than SLCs associated with stages VII-VIII. However, when the SLCs were isolated from each of the two groups of tubules and cultured in vitro, no differences were seen in their ability to proliferate or differentiate. These results suggested that the stage-dependent local factors, not the SLCs themselves, explain the stage-dependent differences in SLC function. TGFB, produced in stage-specific fashion by Sertoli cells, is among the factors shown in previous studies to affect SLC function in vitro. When TGFB inhibitors were included in the cultures of stages IX-XI and VII-VIII tubules, stage-dependent differences in SLC development were reduced, suggesting that TGFB may be among the paracrine factors involved in the stage-dependent differences in SLC function. Taken together, the findings suggest that there is dynamic interaction between SLCs and germ/Sertoli cells within the seminiferous tubules that may affect SLC proliferation and differentiation.


Assuntos
Células Intersticiais do Testículo/citologia , Túbulos Seminíferos/citologia , Células-Tronco/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Células Intersticiais do Testículo/metabolismo , Masculino , Comunicação Parácrina , Ratos , Túbulos Seminíferos/metabolismo , Espermatogênese , Células-Tronco/metabolismo
16.
BMC Pharmacol Toxicol ; 20(1): 32, 2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-31133074

RESUMO

BACKGROUND: It is an established fact that excess of glucocorticoids could cause the harmful effects, such as suppression on the male reproduction. Although glucocorticoids pharmacologically inhibit the Leydig cell function, their roles in Leydig cell development are unclear. Therefore, the present study was designed to investigate effects of synthetic glucocorticoid dexamethasone (DEX) on rat stem Leydig cell proliferation and differentiation. METHODS: Male Sprague-Dawley rats received a single intraperitoneal injection of 75 mg/kg EDS to eliminate Leydig cells and an in vitro culture system of the seminiferous tubules was established from Leydig cell-depleted testis. Using basal medium and Leydig cell differentiation-inducing medium (LIM) in the culture system, we examined the effects of DEX (0-100 nM) on the proliferation and differentiation of the stem Leydig cells in vitro, respectively. RESULTS: Results showed that LIM is a good agent to induce stem Leydig cell differentiation into Leydig cells that produce testosterone in vitro. DEX inhibited the differentiation of stem Leydig cells by reducing the expression levels of Cyp17a1 and Scarb1 and that NR3C1 antagonist RU38486 reversed the DEX-mediated effects. However, DEX are not involved with the proliferation of stem Leydig cells. CONCLUSIONS: DEX suppressed the differentiation of rat Leydig cells in vitro and glucocorticoid-induced effects acted through NR3C1. This suppression partially targets on Cyp17a1 and Scarb1 gene expression.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dexametasona/toxicidade , Glucocorticoides/toxicidade , Células Intersticiais do Testículo/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Expressão Gênica/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Masculino , Ratos Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Células-Tronco/metabolismo , Esteroide 17-alfa-Hidroxilase/genética , Esteroide 17-alfa-Hidroxilase/metabolismo
17.
Mol Cell Endocrinol ; 493: 110449, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31102608

RESUMO

It was reported previously that adult mouse stem Leydig cells (SLCs) express CD51 (integrin α-chain V). However, it is still unclear whether all CD51+ cells are SLCs. In the present study, we found that CD51+ cells can be classified into two sub-groups, a weakly-staining group (CD51+) and a strongly-staining group (CD51++). The CD51+ cells expressed common SLC marker genes, including Nestin, Pdgfra and Coup-tf2, while CD51++ cells did not express these genes. Instead, they expressed macrophage markers, such as F4/80, Cd115 and Tnfa. When these cells were induced to differentiate in vitro, the CD51+ cells, but not CD51++ cells, formed Leydig cells. Overall, our results showed that although SLCs expressed CD51, not all CD51-expressing cells are SLCs. The cells that expressed high levels of CD51 are actually macrophages.


Assuntos
Integrina alfaV/metabolismo , Células Intersticiais do Testículo/citologia , Células-Tronco/imunologia , Testículo/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Células Cultivadas , Células Intersticiais do Testículo/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Espermatogênese , Células-Tronco/citologia , Testículo/imunologia , Regulação para Cima
18.
Environ Pollut ; 250: 206-215, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30999198

RESUMO

Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic (PFOS) are two perfluorinated chemical products widely existing in the environment. Evidence suggested that PFOA might relate to male reproductive dysfunction in rats and humans. PFOA exposure inhibited the function of Leydig cells. However, it is still unknown whether PFOA affects stem Leydig cells (SLCs). In the present study, we examined the effects of a short-term exposure to PFOA on Leydig cell regeneration and also explored the possible mechanism involved. Thirty-six adult Sprague-Dawley rats were randomly divided into three groups and intraperitoneally injected with a single dose of 75 mg/kg ethane dimethyl sulfonate (EDS) to eliminate all Leydig cells. From post-EDS day 7, the 3 group rats received 0, 25 or 50 mg/kg/day PFOA (n = 12 per group) for 9 consecutive days. Exposure to PFOA significantly decreased serum testosterone levels by day 21 and day 56 post-EDS treatment. Also, the expression levels of Leydig cell specific genes (Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Hsd11b1 and Cyp17a1) and their protein levels were all down-regulated. PFOA exposure may also affect proliferation of SLCs or their progeny since the numbers of PCNA-positive Leydig cells were reduced by post-EDS day 21. These in vivo observations were also confirmed by in vitro studies where the effects of PFOA were tested by culture of seminiferous tubules. In summary, PFOA exposure inhibits the development of Leydig cells, possibly by affecting both the proliferation and differentiation of SLCs or their progeny.


Assuntos
Caprilatos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Células Intersticiais do Testículo/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/metabolismo , Masculino , Ratos Sprague-Dawley , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Testosterona/sangue
19.
Stem Cell Res ; 41: 101593, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31704538

RESUMO

Adult testicular Leydig cells arise from stem cells in the neonatal and adult testis. The nature of these stem Leydig cells (SLCs) have not been well characterized. We have found previously that a group cells expressing CD90, a cell surface glycoprotein that may play roles in cell-cell and cell-matrix interactions and associated with the seminiferous tubule surface, have the ability to form Leydig cells. As yet, the relationship between this CD90+ cell population and SLCs reported previously by other groups is still unknown. In the present study, we systematically characterized these CD90+ cells by their ability to express multiple potential SLC markers and to proliferate and differentiate into Leydig cells in vitro. First, we have found by qPCR and immunohistochemical staining that the CD90+ cells do not express any of the markers of the common seminiferous tubular cells, including myoid, Sertoli, germ and Leydig cells, as well as macrophages. Moreover, when the CD90+ cells were isolated by fluorescent-sorting, the cells expressed high levels of all the potential SLC marker genes, including Nestin, Cd51, Coup-tf2, Arx, Pdgfra and Tcf21. Also, CD90-positive, but not -negative, cells were able to form Leydig cells in vitro with the proper inducing medium. Overall, the results indicated that the tubule-associated CD90+ cells represent a population of SLC in adult testis.


Assuntos
Células-Tronco Adultas/metabolismo , Antígenos de Diferenciação/metabolismo , Células Intersticiais do Testículo/metabolismo , Túbulos Seminíferos/metabolismo , Células-Tronco Adultas/citologia , Animais , Células Intersticiais do Testículo/citologia , Masculino , Ratos , Ratos Sprague-Dawley , Túbulos Seminíferos/citologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-29416526

RESUMO

Aldosterone (ALDO) is a primary endogenous mineralocorticoid, appearing as the main hormone controlling sodium and water homeostasis. Its emerging role in the development of many organs has gained interest over the past few years. In the testis, Leydig cells contain mineralocorticoid receptors and ALDO stimulates androgen synthesis via the mineralocorticoid receptors in rat adult Leydig cells. Although ALDO pharmacologically promoted the Leydig cell function, its role in Leydig cell development was unclear. In the present study, we investigated effects of ALDO on rat stem Leydig cell (SLC) proliferation and differentiation. Using an in vitro culture system of the seminiferous tubules from Leydig cell-depleted testis and EdU (a modified thymidine analog) incorporation into the SLC for flurorescent labeling to judge its DNA synthesis and measurement of medium testosterone production, steroidogenesis-related gene and protein expression, we found that: (1) ALDO suppressed EdU incorporation into SLCs at 100 nM via mineralocorticoid receptor-mediated mechanism and (2) ALDO reduced Leydig cell number. In conclusion, ALDO pharmacologically blocked rat SLC development.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA