Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
EXCLI J ; 19: 1459-1476, 2020.
Article in English | MEDLINE | ID: mdl-33312107

ABSTRACT

The debate about possible adverse effects of bisphenol A (BPA) has been ongoing for decades. Bisphenol F (BPF) and S (BPS) have been suggested as "safer" alternatives. In the present study we used hepatocyte-like cells (HLCs) derived from the human embryonic stem cell lines Man12 and H9 to compare the three bisphenol derivatives. Stem cell-derived progenitors were produced using an established system and were exposed to BPA, BPF and BPS for 8 days during their transition to HLCs. Subsequently, we examined cell viability, inhibition of cytochrome P450 (CYP) activity, and genome-wide RNA profiles. Sub-cytotoxic, inhibitory concentrations (IC50) of CYP3A were 20, 9.5 and 25 µM for BPA, BPF and BPS in Man12 derived HLCs, respectively. The corresponding concentrations for H9-derived HLCs were 19, 29 and 31 µM. These IC50 concentrations were used to study global expression changes in this in vitro study and are higher than unconjugated BPA in serum of the general population. A large overlap of up- as well as downregulated genes induced by the three bisphenol derivatives was seen. This is at least 28-fold higher compared to randomly expected gene expression changes. Moreover, highly significant correlations of expression changes induced by the three bisphenol derivatives were obtained in pairwise comparisons. Dysregulated genes were associated with reduced metabolic function, cellular differentiation, embryonic development, cell survival and apoptosis. In conclusion, no major differences in cytochrome inhibitory activities of BPA, BPF and BPS were observed and gene expression changes showed a high degree of similarity.

2.
BMC Genomics ; 18(1): 972, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29246116

ABSTRACT

BACKGROUND: The Sertoli cells act to induce testis differentiation and subsequent development in fetal and post-natal life which makes them key to an understanding of testis biology. As a major step towards characterisation of factors involved in Sertoli cell function we have identified Sertoli cell-specific transcripts in the mouse testis and have used the data to identify Sertoli cell-specific transcripts altered in mice lacking follicle-stimulating hormone receptors (FSHRKO) and/or androgen receptors (AR) in the Sertoli cells (SCARKO). RESULTS: Adult iDTR mice were injected with busulfan to ablate the germ cells and 50 days later they were treated with diphtheria toxin (DTX) to ablate the Sertoli cells. RNAseq carried out on testes from control, busulfan-treated and busulfan + DTX-treated mice identified 701 Sertoli-specific transcripts and 4302 germ cell-specific transcripts. This data was mapped against results from microarrays using testicular mRNA from 20 day-old FSHRKO, SCARKO and FSHRKO.SCARKO mice. Results show that of the 534 Sertoli cell-specific transcripts present on the gene chips, 85% were altered in the FSHRKO mice and 94% in the SCARKO mice (mostly reduced in both cases). In the FSHRKO.SCARKO mice additive or synergistic effects were seen for most transcripts. Age-dependent studies on a selected number of Sertoli cell-specific transcripts, showed that the marked effects in the FSHRKO at 20 days had largely disappeared by adulthood although synergistic effects of FSHR and AR knockout were seen. CONCLUSIONS: These studies have identified the Sertoli cell-specific transcriptome in the mouse testis and have shown that most genes in the transcriptome are FSH- and androgen-dependent at puberty although the importance of FSH diminishes towards adulthood.


Subject(s)
Receptors, Androgen/genetics , Receptors, FSH/genetics , Sertoli Cells/metabolism , Testis/metabolism , Androgens/physiology , Animals , Busulfan/pharmacology , Diphtheria Toxin/pharmacology , Follicle Stimulating Hormone/physiology , Male , Mice , Mice, Knockout , Spermatozoa/metabolism , Testis/drug effects , Transcriptome/drug effects
3.
Andrology ; 3(6): 1035-49, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26446427

ABSTRACT

Testicular development and function is the culmination of a complex process of autocrine, paracrine and endocrine interactions between multiple cell types. Dissecting this has classically involved the use of systemic treatments to perturb endocrine function, or more recently, transgenic models to knockout individual genes. However, targeting genes one at a time does not capture the more wide-ranging role of each cell type in its entirety. An often overlooked, but extremely powerful approach to elucidate cellular function is the use of cell ablation strategies, specifically removing one cellular population and examining the resultant impacts on development and function. Cell ablation studies reveal a more holistic overview of cell-cell interactions. This not only identifies important roles for the ablated cell type, which warrant further downstream study, but also, and importantly, reveals functions within the tissue that occur completely independently of the ablated cell type. To date, cell ablation studies in the testis have specifically removed germ cells, Leydig cells, macrophages and recently Sertoli cells. These studies have provided great leaps in understanding not possible via other approaches; as such, cell ablation represents an essential component in the researchers' tool-kit, and should be viewed as a complement to the more mainstream approaches to advancing our understanding of testis biology. In this review, we summarise the cell ablation models used in the testis, and discuss what each of these have taught us about testis development and function.


Subject(s)
Ablation Techniques , Leydig Cells/pathology , Macrophages/pathology , Sertoli Cells/pathology , Spermatozoa/pathology , Testis/pathology , Animals , Cell Communication , Humans , Leydig Cells/drug effects , Leydig Cells/metabolism , Leydig Cells/radiation effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/radiation effects , Male , Models, Animal , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sertoli Cells/radiation effects , Signal Transduction , Spermatogenesis , Spermatozoa/drug effects , Spermatozoa/metabolism , Spermatozoa/radiation effects , Testis/drug effects , Testis/metabolism , Testis/radiation effects
4.
Reproduction ; 147(5): 671-82, 2014 May.
Article in English | MEDLINE | ID: mdl-24505118

ABSTRACT

The adult population of Leydig cells acts to secrete testosterone which is essential for reproductive health and fertility in the adult male. However, other physiological functions of these cells are uncertain, and to address this issue a cell ablation model has been used to identify Leydig cell-specific mRNA transcripts. Ethane dimethane sulphonate (EDS) was synthesised by a novel process and was used to ablate Leydig cells in adult male rats previously treated with butane dimethane sulphonate (busulphan) to delete the germ cell population. Levels of mRNA transcripts were measured in the testis using microarrays 1, 3, 5, 8 and 12 days after EDS injection. During this period, there was a significant change in the levels of 2200 different transcripts with a marked decline in the levels of canonical Leydig cell transcripts, such as Cyp11a1, Cyp17a1 and Insl3. A total of 95 transcripts showed a similar decline in expression after EDS treatment, suggesting that they have a Leydig cell-specific origin. Analysis of selected transcripts confirmed that they were expressed specifically in Leydig cells and showed that most had a late onset of expression during adult Leydig cell development. Apart from transcripts encoding components of the steroidogenic apparatus, the most common predicted function of translated proteins was endogenous and xenotoxicant metabolism. In addition, a number of transcripts encode acute-phase proteins involved in reduction of oxidative stress. Results show that, in addition to androgen secretion, Leydig cells may have a critical role to play in protecting the testis from damage caused by toxicants or stress.


Subject(s)
Leydig Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Testis/metabolism , Transcription, Genetic , Animals , Apoptosis/drug effects , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Insulin/genetics , Insulin/metabolism , Leydig Cells/cytology , Leydig Cells/drug effects , Male , Mesylates/pharmacology , Models, Animal , Oxidative Stress/physiology , Proteins/genetics , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testis/cytology , Testis/drug effects
5.
Mol Hum Reprod ; 19(3): 177-87, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23197595

ABSTRACT

The human feto-maternal unit produces large amounts of steroid hormones, particularly estrogens, during the second and third trimesters. The fetal adrenal gland and the placenta are considered the principal tissues driving steroid production but the fetal liver is likely to play an essential role in this process. This study was designed to measure transcript expression of proteins involved in steroid synthesis, metabolism, conjugation and signalling in the human fetal liver and to examine sex differences and effects of maternal smoking. Liver samples were taken from 55 normal fetuses from women undergoing second trimester elective termination. Levels of 23 mRNA transcripts encoding steroid synthesis/metabolic/conjugation enzymes and steroid receptors were measured by real-time PCR. The expression of representative proteins was confirmed by western blotting and immunohistochemistry. The human fetal livers expressed high levels of CYP19A1, SULT2A1, SULT1E1, HSD17B2, SRD5A3 and CYP3A7. Lower levels of SULT1A1, STS, UGT2B17, GPER, AKR1C3, UGT2B15, AR, CYP11A1, CYP21A2, HSD17B3, HSD17B1 and SRD5A1 were also detectable. The expression of ESR, ESR2, CYP17A1 and HSD3B transcripts was undetectable in most fetal livers, although HSD3B was shown to be present by western blotting. Sex differences were limited to SRD5A3 (lower in females) and UGT2B17 (higher in females). Maternal smoking increased the expression of CYP19A1, SULT2A1, UGT2B17, HSD17B2 and AKR1C3 and reduced the expression of SRD5A3 in the male fetal liver. This study shows that the human fetal liver is likely to have an extensive effect on circulating steroid levels in the human fetus and mother. The most important of these effects will be alterations to the species, conjugation and availability of estrogens in the fetus. Maternal smoking is likely to reduce circulating androgen bioactivity in male fetuses.


Subject(s)
Androgens/genetics , Estrogens/genetics , Fetal Proteins/genetics , Fetus/enzymology , Gene Expression Regulation, Developmental , Liver/enzymology , Adult , Androgens/metabolism , Endocrine System/metabolism , Estrogens/metabolism , Female , Fetal Proteins/metabolism , Gene Expression Profiling , Humans , Male , Placenta/enzymology , Pregnancy , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Sex Characteristics , Smoking
6.
Endocrinology ; 151(5): 2343-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20228170

ABSTRACT

Androgens act to stimulate spermatogenesis through androgen receptors (ARs) on the Sertoli cells and peritubular myoid cells. Specific ablation of the AR in either cell type will cause a severe disruption of spermatogenesis. To determine whether androgens can stimulate spermatogenesis through direct action on the peritubular myoid cells alone or whether action on the Sertoli cells is essential, we crossed hypogonadal (hpg) mice that lack gonadotrophins and intratesticular androgen with mice lacking ARs either ubiquitously (ARKO) or specifically on the Sertoli cells (SCARKO). These hpg.ARKO and hpg.SCARKO mice were treated with testosterone (T) or dihydrotestosterone (DHT) for 7 d and testicular morphology and cell numbers assessed. Androgen treatment did not affect Sertoli cell numbers in any animal group. Both T and DHT increased numbers of spermatogonia and spermatocytes in hpg mice, but DHT has no effect on germ cell numbers in hpg.SCARKO and hpg.ARKO mice. T increased germ cell numbers in hpg.SCARKO and hpg.ARKO mice, but this was associated with stimulation of FSH release. Results show that androgen stimulation of spermatogenesis requires direct androgen action on the Sertoli cells.


Subject(s)
Dihydrotestosterone/pharmacology , Sertoli Cells/drug effects , Spermatogenesis/drug effects , Testosterone/pharmacology , Androgens/pharmacology , Animals , Cell Count , Enzyme-Linked Immunosorbent Assay , Female , Follicle Stimulating Hormone/blood , Immunohistochemistry , Male , Mice , Mice, Knockout , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Spermatocytes/cytology , Spermatocytes/drug effects , Spermatocytes/metabolism , Spermatogonia/cytology , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/cytology , Testis/drug effects , Testis/metabolism
7.
Reproduction ; 139(1): 177-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19846485

ABSTRACT

FSH and androgen act to stimulate and maintain spermatogenesis. FSH acts directly on the Sertoli cells to stimulate germ cell number and acts indirectly to increase androgen production by the Leydig cells. In order to differentiate between the direct effects of FSH on spermatogenesis and those mediated indirectly through androgen action, we have crossed hypogonadal (hpg) mice, which lack gonadotrophins, with mice lacking androgen receptors (AR) either ubiquitously (ARKO) or specifically on the Sertoli cells (SCARKO). These hpg.ARKO and hpg.SCARKO mice were treated with recombinant FSH for 7 days and testicular morphology and cell numbers were assessed. In untreated hpg and hpg.SCARKO mice, germ cell development was limited and did not progress beyond the pachytene stage. In hpg.ARKO mice, testes were smaller with fewer Sertoli cells and germ cells compared to hpg mice. Treatment with FSH had no effect on Sertoli cell number but significantly increased germ cell numbers in all groups. In hpg mice, FSH increased the numbers of spermatogonia and spermatocytes, and induced round spermatid formation. In hpg.SCARKO and hpg.ARKO mice, in contrast, only spermatogonial and spermatocyte numbers were increased with no formation of spermatids. Leydig cell numbers were increased by FSH in hpg and hpg.SCARKO mice but not in hpg.ARKO mice. Results show that in rodents 1) FSH acts to stimulate spermatogenesis through an increase in spermatogonial number and subsequent entry of these cells into meiosis, 2) FSH has no direct effect on the completion of meiosis and 3) FSH effects on Leydig cell number are mediated through interstitial ARs.


Subject(s)
Follicle Stimulating Hormone/physiology , Gonadotropins/physiology , Hypogonadism/physiopathology , Receptors, Androgen/physiology , Seminal Vesicles/pathology , Spermatogenesis , Testis/pathology , Animals , Cell Count , Follicle Stimulating Hormone/pharmacology , Gonadotropins/deficiency , Gonadotropins/genetics , Hypogonadism/genetics , Hypogonadism/pathology , Leydig Cells/pathology , Male , Meiosis , Mice , Organ Size , Organ Specificity , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Recombinant Proteins/pharmacology , Sertoli Cells/pathology , Species Specificity , Spermatozoa/pathology , Testis/metabolism , Testosterone/metabolism
8.
Bone Marrow Transplant ; 45(6): 1068-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19915634

ABSTRACT

GVHD is partly mediated by host APCs that activate donor T cells. Extracorporeal photopheresis (ECP) can modulate APC function and benefit some patients with GVHD. We report the results of a study using ECP administered before a standard myeloablative preparative regimen intended to prevent GVHD. Grades II-IV acute GVHD developed in 9 (30%) of 30 recipients of HLA-matched related transplants and 13 (41%) of 32 recipients of HLA-matched unrelated or HLA-mismatched related donor transplants. Actuarial estimates of overall survival (OS) at day 100 and 1-year post transplant were 89% (95% CI, 78-94%) and 77% (95% CI, 64-86%), respectively. There were no unexpected adverse effects of ECP. Historical controls receiving similar conditioning and GVHD prophylaxis regimens but no ECP were identified from the database of the Center for International Blood and Marrow Transplant Research and multivariate analysis indicated a lower risk of grades II-IV acute GVHD in patients receiving ECP (P=0.04). Adjusted OS at 1 year was 83% in the ECP study group and 67% in the historical control group (relative risk 0.44; 95% CI, 0.24-0.80) (P=0.007). These preliminary data may indicate a potential survival advantage with ECP for transplant recipients undergoing standard myeloablative hematopoietic cell transplantation.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Photopheresis/methods , Transplantation Conditioning/adverse effects , Acute Disease , Adolescent , Adult , Female , HLA Antigens , Hematologic Neoplasms/complications , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Middle Aged , Myeloablative Agonists/adverse effects , Survival Rate , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome , Young Adult
9.
J Mol Endocrinol ; 42(4): 291-303, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19136570

ABSTRACT

FSH acts through the Sertoli cell to ensure normal testicular development and function. To identify transcriptional mechanisms through which FSH acts in the testis, we have treated gonadotrophin-deficient hypogonadal (hpg) mice with recombinant FSH and measured changes in testicular transcript levels using microarrays and real-time PCR 12, 24 and 72 h after the start of treatment. Approximately 400 transcripts were significantly altered at each time point by FSH treatment. At 12 h, there was a clear increase in the levels of a number of known Sertoli cell transcripts (e.g. Fabp5, Lgals1, Tesc, Scara5, Aqp5). Additionally, levels of Leydig cell transcripts were also markedly increased (e.g. Ren1, Cyp17a1, Akr1b7, Star, Nr4a1). This was associated with a small but significant rise in testosterone at 24 and 72 h. At 24 h, androgen-dependent Sertoli cell transcripts were up-regulated (e.g. Rhox5, Drd4, Spinlw1, Tubb3 and Tsx) and this trend continued up to 72 h. By contrast with the somatic cells, only five germ cell transcripts (Dkkl1, Hdc, Pou5f1, Zfp541 and 1700021K02Rik) were altered by FSH within the time-course of the experiment. Analysis of canonical pathways showed that FSH induced a general decline in transcripts related to formation and regulation of tight junctions. Results show that FSH acts directly and indirectly to induce rapid changes in Sertoli cell and Leydig cell transcript levels in the hpg mouse but that effects on germ cell development must occur over a longer time-span.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Hypogonadism/metabolism , Testis/drug effects , Testis/metabolism , Animals , Gene Expression Regulation/drug effects , Germ Cells/drug effects , Germ Cells/metabolism , Humans , Hypogonadism/pathology , Male , Metabolic Networks and Pathways/drug effects , Mice , Oligonucleotide Array Sequence Analysis , Organ Size/drug effects , Organ Specificity/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testis/cytology , Testis/ultrastructure
10.
Mol Cell Endocrinol ; 306(1-2): 2-8, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19059463

ABSTRACT

Development and maintenance of the male phenotype and establishment of fertility are all dependent upon the activity of the Sertoli cells and Leydig cells of the testis. This review examines the regulation and function of these cell during fetal and post-natal development. Fetal Leydig cells are sensitive to both luteinising hormone (LH) and adrenocorticotrophic hormone (ACTH) but Leydig cell function appears normal in fetal mice lacking both hormones or their receptors. Post-natally, the Sertoli cells and Leydig cells are reliant upon the pituitary gonadotrophins. Leydig cells are critically dependent on LH but follicle-stimulating hormone (FSH), presumably acting through the Sertoli cell, can also affect Leydig cell function. Testosterone secreted by the Leydig cells acts with FSH to stimulate Sertoli cell activity and spermatogenesis. Study of animals lacking FSH-receptors and androgen-receptors shows that both hormones can act to maintain the meiotic germ cell population but that androgens are critical for completion of meiosis.


Subject(s)
Androgens/metabolism , Gonadotropins/metabolism , Leydig Cells/metabolism , Sertoli Cells/metabolism , Animals , Leydig Cells/cytology , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Sertoli Cells/cytology
11.
Reproduction ; 135(6): 851-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18502897

ABSTRACT

Leydig cells in the rat testis can be specifically ablated with ethane dimethane sulfonate (EDS) and will subsequently re-generate. In this study, we have characterized Leydig cell re-generation and expression of selected cell-signaling molecules in a germ cell-free model of EDS action. This model offers the advantage that re-generation occurs on a stable background without confounding changes from the regressing and repopulating germ cell population. Adult rats were treated with busulfan to remove the germ cell population and Leydig cells were then ablated with EDS. Testicular testosterone levels declined markedly within 24 h of EDS treatment and started to recover after 8 days. After EDS treatment there were marked declines in levels of Leydig cell-specific mRNA transcripts coding for steroidogenic enzymes cytochrome P450 11a1 (Cyp11a1), cytochrome P450 17a1 (Cyp17a1), 3beta-hydroxysteroid dehydrogenase type 1 (Hsd3b1), 17beta-hydroxysteroid dehydrogenase type 3 (Hsd17b3) and the LH receptor. Levels of all transcripts recovered within 20 days of EDS treatment apart from Hsd17b3, which remained undetectable up to 20 days. Immunohistochemical localization of CYP11A1 during the phase of early Leydig cell re-generation showed that the Leydig cell precursors are spindle-shaped peritubular cells. Studies on factors which may be involved in Leydig cell re-generation showed there were significant but transient increases in platelet-derived growth factor A (Pdgfa), leukemia inhibitory factor (Lif), and neurofilament heavy polypeptide (Nefh) after EDS, while desert hedgehog (Dhh) levels declined sharply but recovered by 3 days. This study shows that the Leydig cell precursors are peritubular cells and that expression of Pdgfa and Lif is increased at the start of the re-generation process when precursor proliferation is likely to be taking place.


Subject(s)
Leydig Cells/physiology , Regeneration/genetics , Signal Transduction/genetics , Stem Cells/cytology , 17-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Animals , Antispermatogenic Agents , Base Sequence , Busulfan , Cholesterol Side-Chain Cleavage Enzyme/genetics , DNA Primers , Gene Expression , Hedgehog Proteins/genetics , Immunohistochemistry , Leukemia Inhibitory Factor/genetics , Male , Mesylates , Models, Animal , Molecular Sequence Data , Neurofilament Proteins/genetics , Platelet-Derived Growth Factor/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, LH/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Steroid 17-alpha-Hydroxylase/genetics , Testis/cytology , Testis/metabolism , Testosterone/analysis
12.
Reproduction ; 135(6): 839-50, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18390686

ABSTRACT

It has been shown that testicular germ cell development is critically dependent upon somatic cell activity but, conversely, the extent to which germ cells normally regulate somatic cell function is less clear. This study was designed, therefore, to examine the effect of germ cell depletion on Sertoli cell and Leydig cell transcript levels. Mice were treated with busulphan to deplete the germ cell population and levels of mRNA transcripts encoding 26 Sertoli cell-specific proteins and 6 Leydig cell proteins were measured by real-time PCR up to 50 days after treatment. Spermatogonia were lost from the testis between 5 and 10 days after treatment, while spermatocytes were depleted after 10 days and spermatids after 20 days. By 30 days after treatment, most tubules were devoid of germ cells. Circulating FSH and intratesticular testosterone were not significantly affected by treatment. Of the 26 Sertoli cell markers tested, 13 showed no change in transcript levels after busulphan treatment, 2 showed decreased levels, 9 showed increased levels and 2 showed a biphasic response. In 60% of cases, changes in transcript levels occurred after the loss of the spermatids. Levels of mRNA transcripts encoding Leydig cell-specific products related to steroidogenesis were unaffected by treatment. Results indicate (1) that germ cells play a major and widespread role in the regulation of Sertoli cell activity, (2) most changes in transcript levels are associated with the loss of spermatids and (3) Leydig cell steroidogenesis is largely unaffected by germ cell ablation.


Subject(s)
Leydig Cells/metabolism , Sertoli Cells/metabolism , Spermatozoa/physiology , Adaptor Proteins, Signal Transducing , Animals , Antispermatogenic Agents , Base Sequence , Busulfan , Cell Count , Chromosomal Proteins, Non-Histone/genetics , DNA Primers/genetics , Endodeoxyribonucleases , Esterases/genetics , Gene Expression , Male , Mice , Molecular Sequence Data , Platelet-Derived Growth Factor/genetics , Proteins/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/cytology , Spermatocytes/cytology , Spermatogonia/cytology , Time
13.
Endocrinology ; 149(7): 3279-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403489

ABSTRACT

Spermatogenesis in the adult male depends on the action of FSH and androgen. Ablation of either hormone has deleterious effects on Sertoli cell function and the progression of germ cells through spermatogenesis. In this study we generated mice lacking both FSH receptors (FSHRKO) and androgen receptors on the Sertoli cell (SCARKO) to examine how FSH and androgen combine to regulate Sertoli cell function and spermatogenesis. Sertoli cell number in FSHRKO-SCARKO mice was reduced by about 50% but was not significantly different from FSHRKO mice. In contrast, total germ cell number in FSHRKO-SCARKO mice was reduced to 2% of control mice (and 20% of SCARKO mice) due to a failure to progress beyond early meiosis. Measurement of Sertoli cell-specific transcript levels showed that about a third were independent of hormonal action on the Sertoli cell, whereas others were predominantly androgen dependent or showed redundant control by FSH and androgen. Results show that FSH and androgen act through redundant, additive, and synergistic regulation of spermatogenesis and Sertoli cell activity. In addition, the Sertoli cell retains a significant capacity for activity, which is independent of direct hormonal regulation.


Subject(s)
Receptors, Androgen/physiology , Receptors, FSH/physiology , Sertoli Cells/metabolism , Spermatogenesis/physiology , Androgens/pharmacology , Animals , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Genotype , Male , Mice , Mice, Knockout , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, FSH/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Spermatogenesis/drug effects , Spermatogenesis/genetics
14.
Reproduction ; 135(2): 253-65, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18239053

ABSTRACT

This study was designed to identify genes that regulate the transition from FSH- to LH-dependent development in the bovine dominant follicle (DF). Serial analysis of gene expression (SAGE) was used to compare the transcriptome of granulosa cells isolated from the most oestrogenic growing cohort follicle (COH), the newly selected DF and its largest subordinate follicle (SF) which is destined for atresia. Follicle diameter, follicular fluid oestradiol (E) and E:progesterone ratio confirmed follicle identity. Results show that there are 93 transcript species differentially expressed in DF granulosa cells, but only 8 of these encode proteins known to be involved in DF development. Most characterised transcripts upregulated in the DF are from tissue development genes that regulate cell differentiation, proliferation, apoptosis, signalling and tissue remodelling. Semiquantitative real-time PCR analysis confirmed seven genes with upregulated (P< or =0.05) mRNA expression in DF compared with both COH and SF granulosa cells. Thus, the new genes identified by SAGE and real-time PCR, which show enhanced mRNA expression in the DF, may regulate proliferation (cyclin D2; CCND2), prevention of apoptosis or DNA damage (growth arrest and DNA damage-inducible, beta; GADD45B), RNA synthesis (splicing factor, arginine/serine rich 9; SFRS9) and unknown processes associated with enhanced steroidogenesis (ovary-specific acidic protein; DQ004742) in granulosa cells of DF at the onset of LH-dependent development. Further studies are required to show whether the expression of identified genes is dysregulated when abnormalities occur during DF selection or subsequent development.


Subject(s)
Gene Expression Profiling , Ovarian Follicle/physiology , RNA, Messenger/analysis , Up-Regulation , Animals , Cattle , Estradiol/metabolism , Expressed Sequence Tags , Female , Follicle Stimulating Hormone/genetics , Granulosa Cells/metabolism , Luteinizing Hormone/genetics , Oligonucleotide Array Sequence Analysis , Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
J Clin Endocrinol Metab ; 92(12): 4792-801, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848411

ABSTRACT

CONTEXT: Normal fetal testis development is essential for masculinization and subsequent adult fertility. The second trimester is a critical period of human testicular development and masculinization, but there is a paucity of reliable developmental data. OBJECTIVE: The objective of the study was to analyze second-trimester human testicular morphology and function. DESIGN: This was an observational study of second-trimester testis development. SETTING: The study was conducted at the Universities of Glasgow and Aberdeen. PATIENTS/PARTICIPANTS: Testes were collected from 57 morphologically normal fetuses of women undergoing elective termination of normally progressing pregnancies (11-19 wk gestation). MAIN OUTCOME MEASURE(S): Testicular morphology, cell numbers, and quantitative expression of 22 key testicular genes were determined. RESULTS: Sertoli cell and germ cell number increased exponentially throughout the second trimester. Leydig cell number initially increased exponentially but slowed toward 19 wk. Transcripts encoding Sertoli (KITL, FGF9, SOX9, FSHR, WT1) and germ (CKIT, TFAP2C) cell-specific products increased per testis through the second trimester, but expression per cell was static apart from TFAP2C, which declined. Leydig cell transcripts (HSD17B3, CYP11A1, PTC1, CYP17, LHR, INSL3) also remained static per cell. Testicular expression of adrenal transcripts MC2R, CYP11B1, and CYP21 was detectable but unchanged. Expression of other transcripts known or postulated to be involved in testicular development (GATA4, GATA6, CXORF6, WNT2B, WNT4, WNT5A) increased significantly per testis during the second trimester. CONCLUSIONS: The second trimester is essential for the establishment of Sertoli and germ cell numbers. Sertoli and Leydig cells are active throughout the period, but there is no evidence of changing transcript levels.


Subject(s)
Pregnancy Trimester, Second/metabolism , RNA, Messenger/biosynthesis , Testis/cytology , Testis/metabolism , Adult , Cell Count , Female , Gene Expression/physiology , Germ Cells/metabolism , Humans , Leydig Cells/metabolism , Male , Pregnancy , Proteins/metabolism , Sertoli Cells/metabolism , Testis/embryology , Testosterone/metabolism , Transcription Factors
16.
Endocrinology ; 148(6): 2914-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17332066

ABSTRACT

Androgens are essential for the development and maintenance of spermatogenesis, but the underlying mechanisms of androgen action in the testis remain unclear. To help clarify these mechanisms, gene expression was measured in testes of pubertal (20 d old), androgen-insensitive, testicular feminized (Tfm) mice and in normal controls. Using microarrays (Affymetrix chips 430A and 430B), initial data identified a large number of genes down-regulated in the Tfm testis (>4700). These genes were largely of germ cell origin, reflecting the arrest of spermatogenesis that is apparent in the 20-d-old Tfm testis. Subsequent screening in vitro and in silico of this gene set identified 20 genes of a somatic tubular origin that were significantly down-regulated in the Tfm testis and six genes that were significantly up-regulated. Altered expression of these genes was confirmed by real-time PCR, and genes down-regulated in the Tfm testis were shown to be up-regulated in testes of hypogonadal (hpg) mice treated with androgen. In a developmental study using real-time PCR most of the regulated genes showed normal expression during fetal and neonatal development and deviated from control only between 10 and 20 d. In all cases, expression was also reduced in the adult, although interpretation is more complex because of the inherent cryptorchidism in the adult Tfm mouse. Of the total number of somatic genes showing differential expression in the Tfm testis, 50% were associated with three separate groups of genes involved in regulation of vitamin A metabolism, solute transportation, and cytoskeletal function. Thus, effects of androgens on tubular function and spermatogenesis may be mediated in part through regulation of the tubular environment and control of retinoic acid concentrations.


Subject(s)
Androgen-Insensitivity Syndrome/genetics , Carrier Proteins/metabolism , Cytoskeleton/physiology , Gene Expression Regulation, Developmental , Testis/metabolism , Vitamin A/metabolism , Androgen-Insensitivity Syndrome/pathology , Animals , Carrier Proteins/genetics , Cytoskeleton/genetics , Gene Expression Profiling , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Seminiferous Tubules/metabolism , Spermatogenesis/genetics
17.
Int J Androl ; 29(1): 90-5; discussion 105-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16466528

ABSTRACT

The foetal Leydig cell population arises shortly after testicular differentiation at around 12.5 dpc in the mouse and 6 weeks in the human. These cells function, primarily, to produce androgens which are essential for masculinization of the foetus. The origin of the foetal Leydig cells remains uncertain but it has been suggested that adrenocortical cells and foetal Leydig cells may share a common origin in an adreno-genital primordium. Studies in the mouse are beginning to identify factors such as desert hedgehog and platelet-derived growth factor which are required for foetal Leydig cell development. Regulation of foetal Leydig cell function remains uncertain in most species. Unlike the adult population of Leydig cells, the foetal Leydig cells in the mouse do not require luteinizing hormone (LH) to stimulate androgen production. An intact pituitary does appear to be required, however, and adrenocorticotrophic hormone (ACTH) will stimulate foetal Leydig cell function directly suggesting that both LH and ACTH act to maintain Leydig cell function in vivo. In the human LH/hCG is required for foetal Leydig cell function although the cells may also be sensitive to ACTH.


Subject(s)
Cell Differentiation , Leydig Cells/cytology , Leydig Cells/metabolism , Testis/cytology , Testis/embryology , Animals , Fetus , Humans , Leydig Cells/drug effects , Leydig Cells/physiology , Luteinizing Hormone/pharmacology , Luteinizing Hormone/physiology , Male , Testis/drug effects , Testis/metabolism , Testis/physiology
18.
Ann N Y Acad Sci ; 1061: 109-19, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16467262

ABSTRACT

During development in the mouse, two populations of Leydig cells arise sequentially. The fetal Leydig cell population arises shortly after testicular differentiation and functions primarily to produce androgens that are essential for masculinization of the fetus. The origin of the fetal Leydig stem cells remains uncertain, but it has been suggested that adrenocortical cells and fetal Leydig cells may share a common origin in an adrenogenital primordium. The fetal Leydig cells require an intact pituitary for normal development and are sensitive to both luteinizing hormone (LH) and adrenocorticotrophic hormone (ACTH). Loss of either one of these hormones does not, however, affect fetal androgen production, suggesting that both LH and ACTH may act to maintain fetal Leydig cell function in vivo in a redundant fashion. The adult Leydig cell population starts to develop soon after birth in the mouse. Initial differentiation does not appear to require gonadotropin input, but subsequent development and function are completely dependent upon LH. The adult Leydig cells do not require circulating follicle-stimulating hormone, provided that LH is present, but androgen stimulation, through the androgen receptor, is required for normal Leydig cell development in the mouse. It is likely that the effects of androgen are mediated directly in the Leydig cells or indirectly through the peritubular cells.


Subject(s)
Leydig Cells/physiology , Neurosecretory Systems/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Androgens/metabolism , Animals , Cell Differentiation , Follicle Stimulating Hormone/metabolism , Leydig Cells/cytology , Luteinizing Hormone/metabolism , Male , Mice , Models, Biological , Neurosecretory Systems/cytology , Neurosecretory Systems/embryology , Testis/cytology , Testis/embryology , Testis/physiology
19.
Endocrinology ; 146(1): 309-17, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15459113

ABSTRACT

Changes in gene expression during granulosa cell luteinization have been measured using serial analysis of gene expression (SAGE). Immature normal mice were treated with pregnant mare serum gonadotropin (PMSG) or PMSG followed, 48 h later, by human chorionic gonadotropin (hCG). Granulosa cells were collected from preovulatory follicles after PMSG injection or PMSG/hCG injection and SAGE libraries generated from the isolated mRNA. The combined libraries contained 105,224 tags representing 40,248 unique transcripts. Overall, 715 transcripts showed a significant difference in abundance between the two libraries of which 216 were significantly down-regulated by hCG and 499 were significantly up-regulated. Among transcripts differentially regulated, there were clear and expected changes in genes involved in steroidogenesis as well as clusters of genes involved in modeling of the extracellular matrix, regulation of the cytoskeleton and intra and intercellular signaling. The SAGE libraries described here provide a base for functional investigation of the regulation of granulosa cell luteinization.


Subject(s)
Gene Expression/physiology , Granulosa Cells/physiology , Luteinization/physiology , Animals , Chorionic Gonadotropin/pharmacology , Computer Systems , Female , Gene Expression Profiling , Gonadotropins, Equine/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Mice, Inbred C3H , Polymerase Chain Reaction , Time Factors
20.
Endocrinology ; 144(8): 3279-84, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12865302

ABSTRACT

Adult Leydig cell steroidogenesis is dependent on LH but fetal Leydig cells can function independently of gonadotropin stimulation. To identify factors that may be involved in regulation of fetal Leydig cells expressed sequence tag libraries from fetal and adult testes were compared, and fetal-specific genes identified. The ACTH receptor [melanocortin type 2 receptor (Mc2r)] was identified within this fetal-specific group. Subsequent real-time PCR studies confirmed that Mc2r was expressed in the fetal testis at 100-fold higher levels than in the adult testis. Incubation of fetal or neonatal testes with ACTH in vitro stimulated testosterone production more than 10-fold, although ACTH had no effect on testes from animals aged 20 d or older. The steroidogenic response of fetal and neonatal testes to a maximally stimulating dose of human chorionic gonadotropin was similar to the response shown to ACTH. The ED(50) for ACTH, measured in isolated fetal and neonatal testicular cells, was 5 x 10(-10) M and the lowest dose of ACTH eliciting a response was 2 x 10(-11) M. Circulating ACTH levels in fetal mice were around 8 x 10(-11) M. Neither alpha-MSH nor gamma-MSH had any effect on androgen production in vitro at any age. Fetal testosterone levels were normal in mice that lack circulating ACTH (proopiomelanocortin-null) indicating that ACTH is not essential for fetal Leydig cell function. Results show that both LH and ACTH can regulate testicular steroidogenesis during fetal development in the mouse and suggest that fetal Leydig cells, but not adult Leydig cells, are sensitive to ACTH stimulation.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Animals, Newborn/metabolism , Testis/embryology , Testis/metabolism , Testosterone/biosynthesis , Adrenocorticotropic Hormone/blood , Animals , Chorionic Gonadotropin/pharmacology , Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Male , Mice , Mice, Inbred C3H , Mice, Knockout , Polymerase Chain Reaction , Pro-Opiomelanocortin/deficiency , Receptor, Melanocortin, Type 2 , Receptors, Corticotropin/analysis , Receptors, Corticotropin/genetics , Testis/drug effects , Testosterone/analysis , alpha-MSH/pharmacology , gamma-MSH/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...