scholarly journals Distinct Roles for Steroidogenic factor 1 and Desert hedgehog Pathways in Fetal and Adult Leydig Cell Development

Endocrinology ◽  
2007 ◽  
Vol 148 (8) ◽  
pp. 3704-3710 ◽  
Author(s):  
Susan Y. Park ◽  
Minghan Tong ◽  
J. Larry Jameson

Testicular Leydig cells produce testosterone and provide the hormonal environment required for male virilization and spermatogenesis. In utero, fetal Leydig cells (FLCs) are necessary for the development of the Wolffian duct and male external genitalia. Steroidogenic factor 1 (Sf1) is a transcriptional regulator of hormone biosynthesis genes, thus serving a central role in the Leydig cell. Desert hedgehog (Dhh), a Sertoli cell product, specifies the FLC lineage in the primordial gonad through a paracrine signaling mechanism. Postnatally, FLCs are replaced in the testis by morphologically distinct adult Leydig cells (ALCs). To study a putative interaction between Sf1 and Dhh, we crossed Sf1 heterozygous mutant mice with Dhh homozygous null mice to test the function of these two genes in vivo. All of the compound Sf1+/−; Dhh−/− mutants failed to masculinize and were externally female. However, embryonic gonads contained anastomotic testis cords with Sertoli cells and germ cells, indicating that sex reversal was not attributable to a fate switch of the early gonad. Instead, external feminization was attributable to the absence of differentiated FLCs in XY compound mutant mice. ALCs also failed to develop, suggesting either a dependence of ALCs on the prenatal establishment of Leydig cell precursors or that Sf1 and Dhh are both required for ALC maturation. In summary, this study provides genetic evidence that combinatorial expression of the paracrine factor Dhh and nuclear transcription factor Sf1 is required for Leydig cell development.

2019 ◽  
Vol 34 (9) ◽  
pp. 1621-1631 ◽  
Author(s):  
J Eliveld ◽  
E A van den Berg ◽  
J V Chikhovskaya ◽  
S K M van Daalen ◽  
C M de Winter-Korver ◽  
...  

Abstract STUDY QUESTION Is it possible to differentiate primary human testicular platelet-derived growth factor receptor alpha positive (PDGFRα+) cells into functional Leydig cells? SUMMARY ANSWER Although human testicular PDGFRα+ cells are multipotent and are capable of differentiating into steroidogenic cells with Leydig cell characteristics, they are not able to produce testosterone after differentiation. WHAT IS KNOWN ALREADY In rodents, stem Leydig cells (SLCs) that have been identified and isolated using the marker PDGFRα can give rise to adult testosterone-producing Leydig cells after appropriate differentiation in vitro. Although PDGFRα+ cells have also been identified in human testicular tissue, so far there is no evidence that these cells are true human SLCs that can differentiate into functional Leydig cells in vitro or in vivo. STUDY DESIGN, SIZE, DURATION We isolated testicular cells enriched for interstitial cells from frozen–thawed fragments of testicular tissue from four human donors. Depending on the obtained cell number, PDGFRα+-sorted cells of three to four donors were exposed to differentiation conditions in vitro to stimulate development into adipocytes, osteocytes, chondrocytes or into Leydig cells. We compared their cell characteristics with cells directly after sorting and cells in propagation conditions. To investigate their differentiation potential in vivo, PDGFRα+-sorted cells were transplanted in the testis of 12 luteinizing hormone receptor-knockout (LuRKO) mice of which 6 mice received immunosuppression treatment. An additional six mice did not receive cell transplantation and were used as a control. PARTICIPANTS/MATERIALS, SETTING, METHODS Human testicular interstitial cells were cultured to Passage 3 and FACS sorted for HLA-A,B,C+/CD34−/PDGFRα+. We examined their mesenchymal stromal cell (MSC) membrane protein expression by FACS analyses. Furthermore, we investigated lineage-specific staining and gene expression after MSC trilineage differentiation. For the differentiation into Leydig cells, PDGFRα+-sorted cells were cultured in either proliferation or differentiation medium for 28 days, after which they were stimulated either with or without hCG, forskolin or dbcAMP for 24 h to examine the increase in gene expression of steroidogenic enzymes using qPCR. In addition, testosterone, androstenedione and progesterone levels were measured in the culture medium. We also transplanted human PDGFRα+-sorted testicular interstitial cells into the testis of LuRKO mice. Serum was collected at several time points after transplantation, and testosterone was measured. Twenty weeks after transplantation testes were collected for histological examination. MAIN RESULTS AND THE ROLE OF CHANCE From primary cultured human testicular interstitial cells at Passage 3, we could obtain a population of HLA-A,B,C+/CD34−/PDGFRα+ cells by FACS. The sorted cells showed characteristics of MSC and were able to differentiate into adipocytes, chondrocytes and osteocytes. Upon directed differentiation into Leydig cells in vitro, we observed a significant increase in the expression of HSD3B2 and INSL3. After 24 h stimulation with forskolin or dbcAMP, a significantly increased expression of STAR and CYP11A1 was observed. The cells already expressed HSD17B3 and CYP17A1 before differentiation but the expression of these genes were not significantly increased after differentiation and stimulation. Testosterone levels could not be detected in the medium in any of the stimulation conditions, but after stimulation with forskolin or dbcAMP, androstenedione and progesterone were detected in culture medium. After transplantation of the human cells into the testes of LuRKO mice, no significant increase in serum testosterone levels was found compared to the controls. Also, no human cells were identified in the interstitium of mice testes 20 weeks after transplantation. LARGE SCALE DATA N/A LIMITATIONS, REASONS FOR CAUTION This study was performed using tissue from only four donors because of limitations in donor material. Because of the need of sufficient cell numbers, we first propagated cells to passage 3 before FACS of the desired cell population was performed. We cannot rule out this propagation of the cells resulted in loss of stem cell properties. WIDER IMPLICATIONS OF THE FINDINGS A lot of information on Leydig cell development is obtained from rodent studies, while the knowledge on human Leydig cell development is very limited. Our study shows that human testicular interstitial PDGFRα+ cells have different characteristics compared to rodent testicular PDGFRα+ cells in gene expression levels of steroidogenic enzymes and potential to differentiate in adult Leydig cells under comparable culture conditions. This emphasizes the need for confirming results from rodent studies in the human situation to be able to translate this knowledge to the human conditions, to eventually contribute to improvements of testosterone replacement therapies or establishing alternative cell therapies in the future, potentially based on SLCs. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by Amsterdam UMC, location AMC, Amsterdam, the Netherlands. All authors declare no competing interests.


2005 ◽  
Vol 187 (1) ◽  
pp. 117-124 ◽  
Author(s):  
K Svechnikov ◽  
V Supornsilchai ◽  
M-L Strand ◽  
A Wahlgren ◽  
D Seidlova-Wuttke ◽  
...  

Procymidone is a fungicide with anti-androgenic properties, widely used to protect fruits from fungal infection. Thereby it contaminates fruit products prepared for human consumption. Genistein-containing soy products are increasingly used as food additives with health-promoting properties. Therefore we examined the effects of long-term dietary administration (3 months) of the anti-androgen procymidone (26.4 mg/animal per day) or the phytoestrogen genistein (21.1 mg/animal per day) to rats on the pituitary-gonadal axis in vivo, as well as on Leydig cell steroidogenesis and on spermatogenesis ex vivo. The procymidone-containing diet elevated serum levels of LH and testosterone and, furthermore, Leydig cells isolated from procymidone-treated animals displayed an enhanced capacity for producing testosterone in response to stimulation by hCG or dibutyryl cAMP, as well as elevated expression of steroidogenic acute regulatory protein (StAR), cytochrome P450 side-chain cleavage (P450 scc) and cytochrome P450 17α (P450c17). In contrast, the rate of DNA synthesis during stages VIII and IX of spermatogenesis in segments of seminiferous tubules isolated from genistein-treated rats was decreased without accompanying changes in the serum level of either LH or testosterone. Nonetheless, genistein did suppress the ex vivo steroidogenic response of Leydig cells to hCG or dibutyryl cAMP by down-regulating their expression of P450 scc. Considered together, our present findings demonstrate that long-term dietary administration of procymidone or genistein to rats exerts different effects on the pituitary–gonadal axis in vivo and on Leydig cell steroidogenesis ex vivo. Possibly as a result of disruption of hormonal feedback control due to its anti-androgenic action, procymidone activates this endocrine axis, thereby causing hyper-gonadotropic activation of testicular steroidogenesis. In contrast, genistein influences spermatogenesis and significantly inhibits Leydig cell steroidogenesis ex vivo without altering the serum level of either LH or testosterone.


Reproduction ◽  
2017 ◽  
Vol 154 (4) ◽  
pp. 455-467 ◽  
Author(s):  
Gervette M Penny ◽  
Rebecca B Cochran ◽  
Marjut Pihlajoki ◽  
Antti Kyrönlahti ◽  
Anja Schrade ◽  
...  

Testicular Leydig cells produce androgens essential for proper male reproductive development and fertility. Here, we describe a new Leydig cell ablation model based on Cre/Lox recombination of mouse Gata4 and Gata6, two genes implicated in the transcriptional regulation of steroidogenesis. The testicular interstitium of adult Gata4flox/flox; Gata6flox/flox mice was injected with adenoviral vectors encoding Cre + GFP (Ad-Cre-IRES-GFP) or GFP alone (Ad-GFP). The vectors efficiently and selectively transduced Leydig cells, as evidenced by GFP reporter expression. Three days after Ad-Cre-IRES-GFP injection, expression of androgen biosynthetic genes (Hsd3b1, Cyp17a1 and Hsd17b3) was reduced, whereas expression of another Leydig cell marker, Insl3, was unchanged. Six days after Ad-Cre-IRES-GFP treatment, the testicular interstitium was devoid of Leydig cells, and there was a concomitant loss of all Leydig cell markers. Chromatin condensation, nuclear fragmentation, mitochondrial swelling, and other ultrastructural changes were evident in the degenerating Leydig cells. Liquid chromatography-tandem mass spectrometry demonstrated reduced levels of androstenedione and testosterone in testes from mice injected with Ad-Cre-IRES-GFP. Late effects of treatment included testicular atrophy, infertility and the accumulation of lymphoid cells in the testicular interstitium. We conclude that adenoviral-mediated gene delivery is an expeditious way to probe Leydig cell function in vivo. Our findings reinforce the notion that GATA factors are key regulators of steroidogenesis and testicular somatic cell survival. Free Finnish abstract: A Finnish translation of this abstract is freely available at http://www.reproduction-online.org/content/154/4/455/suppl/DC2.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1533-1533
Author(s):  
David T Yang ◽  
Shelly Wuerzberger-Davis ◽  
Yuhong Chen ◽  
Mei Yu ◽  
Hu Zeng ◽  
...  

Abstract Activity of the nuclear factor-κB (NF-κB) family of transcription factors is tightly regulated by its inhibitor, IκBα, through cytoplasmic localization of latent NF-κB: IκBα complexes. This arrangement is essential for efficient signal-inducible activation and regulation of biologic functions. Maintenance of cytoplasmic localization of latent NF-κB: IκBα complex requires continuous nuclear export that is dependent on the N-terminal nuclear export sequence (N-NES) of IκBα. While these mechanisms have been elucidated through in vitro studies, the biological significance of this “nucleocytoplasmic shuttling” has yet to be evaluated in vivo. To address this, we derived mice harboring germ-line M45A, L48A, and I52A amino acid substitutions in the N-NES of IκBα. In splenic B-cells, the disrupted N-NES caused constitutive nuclear accumulation of IκBα and inactive c-Rel containing complexes but surprisingly not IκBα: p65 complexes. Since p65 contains a NES sequence and c-Rel does not, nuclear export of N-NES mutant IκBα:NF-κB complexes appear to be NF-κB family member dependent. Functionally, NF-κB activity in splenic B-cells after stimulation with IgM or LPS was clearly reduced in the mutants compared to wild-type by electrophoretic mobility shift assay. B-cell development in the bone marrow of mice harboring the mutation was impaired, showing a preponderance of pro/pre B-cells and few mature B-cells compared to their wild type littermates (p < 0.001). Concordantly, there were significantly fewer B-cells in the spleen (p < 0.05) and lymph nodes (p < 0.01) of the mutant mice. Additionally, populations of T2, follicular (FO), and marginal zone (MZ) B-cells, which represent mature B-cells in the spleen, were also reduced in the mutant mice (p < 0.001). To demonstrate that this B-cell maturation defect in IκBα mutant mice was B-cell intrinsic, sublethally irradiated Jak3-deficient mice were transplanted with BM from either wild-type or mutant mice. B-cell development in mice transplanted with mutant donors was impaired relative to those with wild-type donors in a fashion identical to that of the primary mutants described above. Finally, severe phenotypes in inguinal lymph nodes and Peyer’s patch development were present, with mutant mice frequently lacking these secondary organs/tissues, the underlying mechanisms of which are currently being investigated. In conclusion, our findings uncover an in vivo mechanism controlling NF-κB localization and its essential role in the generation of mature B-cells and certain secondary lymphoid organs.


2004 ◽  
Vol 167 (5) ◽  
pp. 935-944 ◽  
Author(s):  
Michail S. Davidoff ◽  
Ralf Middendorff ◽  
Grigori Enikolopov ◽  
Dieter Riethmacher ◽  
Adolf F. Holstein ◽  
...  

The cells responsible for production of the male sex hormone testosterone, the Leydig cells of the testis, are post-mitotic cells with neuroendocrine characteristics. Their origin during ontogeny and regeneration processes is still a matter of debate. Here, we show that cells of testicular blood vessels, namely vascular smooth muscle cells and pericytes, are the progenitors of Leydig cells. Resembling stem cells of the nervous system, the Leydig cell progenitors are characterized by the expression of nestin. Using an in vivo model to induce and monitor the synchronized generation of a completely new Leydig cell population in adult rats, we demonstrate specific proliferation of vascular progenitors and their subsequent transdifferentiation into steroidogenic Leydig cells which, in addition, rapidly acquire neuronal and glial properties. These findings, shown to be representative also for ontogenetic Leydig cell formation and for the human testis, provide further evidence that cellular components of blood vessels can act as progenitor cells for organogenesis and repair.


2015 ◽  
Vol 93 (4) ◽  
Author(s):  
Tatiana Karpova ◽  
Kumarasamy Ravichandiran ◽  
Lovella Insisienmay ◽  
Daren Rice ◽  
Valentine Agbor ◽  
...  

Endocrinology ◽  
2004 ◽  
Vol 145 (11) ◽  
pp. 4825-4834 ◽  
Author(s):  
M. L. Barreiro ◽  
F. Gaytan ◽  
J. M. Castellano ◽  
J. S. Suominen ◽  
J. Roa ◽  
...  

Abstract Ghrelin has emerged as putative regulator of an array of endocrine and nonendocrine functions, including cell proliferation. Recently, we provided evidence for the expression of ghrelin in mature, but not in undifferentiated, Leydig cells of rat and human testis. Yet testicular actions of ghrelin, other than modulation of testosterone secretion, remain unexplored. In the present study we evaluated the effects of ghrelin on proliferation of Leydig cell precursors during puberty and after selective elimination of mature Leydig cells by treatment with ethylene dimethane sulfonate. In these settings, intratesticular injection of ghrelin significantly decreased the proliferative activity of differentiating immature Leydig cells, estimated by 5-bromodeoxyuridine labeling. This response was selective and associated, in ethylene dimethane sulfonate-treated animals, with a decrease in the mRNA levels of stem cell factor (SCF), i.e. a key signal in spermatogenesis and a putative regulator of Leydig cell development. Thus, the effects of ghrelin on SCF gene expression were evaluated. In adult rats, ghrelin induced a significant decrease in SCF mRNA levels in vivo. Such an inhibitory action was also detected in vitro using cultures of staged seminiferous tubules. The inhibitory effect of ghrelin in vivo was dependent on proper FSH input, because it was detected in hypophysectomized rats only after FSH replacement. Overall, it is proposed that acquisition of ghrelin expression by Leydig cell precursors during differentiation may operate as a self-regulatory signal for the inhibition of the proliferative activity of this cell type through direct or indirect (i.e. SCF-mediated) mechanisms. In addition, we present novel evidence for the ability of ghrelin to modulate the expression of the SCF gene, which may have implications for the mode of action of this molecule in the testis as well as in other physiological systems.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Xiangcheng Zhan ◽  
Jingwei Zhang ◽  
Saiyang Li ◽  
Xiaolu Zhang ◽  
Linchao Li ◽  
...  

Abstract 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.


2021 ◽  
Author(s):  
Zhe Cao ◽  
Jun Qiang ◽  
Jun Zhu ◽  
Hong Li ◽  
Yi Tao ◽  
...  

Abstract Steroidogenic factor 1 (sf1) is an important regulator of gonad development and function in mammals. However, study of sf1 in fish is limited to cloning and expression and in vitro experiments. Using antisense RNA we knockout transcription of the sf1 gene in Nile tilapia Oreochromis niloticus, and obtain experimental fish in vivo. We demonstrate that antisense RNA can silence sf1 transcription and protein expression, and report suppression of sf1 transcription to affect gonad development and external genitalia formation in Nile tilapia. We also report disfunction of retinal metabolism and fatty acid metabolism to be important causes of weight gain and gonad abnormality with sf1 suppression. The feasibility of using antisense RNA for gene editing in fish is verified, and a new way of studying gene function and performing biological breeding is presented.


Sign in / Sign up

Export Citation Format

Share Document