4 resultados para Sertoli cell

em DigitalCommons@The Texas Medical Center


Relevância:

100.00% 100.00%

Publicador:

Resumo:

Steroid hormones regulate target cell function via quantitative and qualitative changes in RNA and protein synthesis. In the testis, androgens are known to play an important role in the regulation of spermatogenesis. The Sertoli cell (SC), whose function is thought to be supportive to the developing germ cell, has been implicated as an androgen target cell. Although cytoplasmic androgen receptors and chromatin acceptor sites for androgen-receptor complexes have been found in SC, effects on RNA synthesis have not previously been demonstrated. In this study, SC RNA synthetic activity was characterized and the effect of testosterone on SC nuclear transcriptional activity in vitro assessed. SC exhibited two fold increases in RNA and ribonucleotide pool concentrations during sexual maturation. These changes appeared to correlate with a previously observed increase in protein concentration per cell over an age span of 15-60 days. Following incubation with ('3)H-uridine, SC from older animals incorporated more label into RNA than SC from younger animals. Since the relative concentration of cytidine nucleotides was higher in SC from older rats, the age-related increase in tritium incorporation may reflect an associated increase in incorporation of ('3)H-CMP into RNA. Alternatively, the enhanced labeling may be the result of either a change in the base composition of the RNA resulting in a higher proportion of CMP and UMP in the RNA, or compartmentalization of the nucleotide pools. The physiologic consequences of these maturational alterations of nucleotide pools remains to be elucidated. RNA polymerase activities were characterized in intact nuclei obtained from cultured rat SC. (alpha)-Amanitin resistant RNA polymerase I+III activity was identical when measured in low or high ionic strength (0.05 M or 0.25 M ammonium sulfate (AS)) in the presence of MnCl(,2) or MgCl(,2), with a divalent cation optimum of 1.6 mM. RNA polymerase II was most active in 0.25 M AS and 1.6 mM MnCl(,2). The apparent Km of RNA polymerase II for UTP was 0.016 mM in 0.05 M AS and 0.037 mM in 0.25 M AS. The apparent Km values for total polymerase activity was 0.008 mM and 0.036 mM at low and high ionic strenghts, respectively. These data indicate that Sertoli cell RNA polymerase activities have catalytic properties characteristic of eukaryotic polymerase activities in general. In the presence of 21 (mu)M testosterone, RNA polymerase II activity increased two fold at 15 minutes, then declined but was still elevated over control values six hours after androgen addition. Polymerase I+III activity was not greatly affected by testosterone. The stimulation of polymerase II measured at 15 minutes was dose-dependent, with a maximum at 0.53 nM and no further stimulation up to 10('-5) M (ED(,50) = 0.25 nM testosterone), and was androgen specific. The results of preliminary RNA isolation and characterization experiments suggested that the synthesis of several species of RNA was enhanced by testosterone administration. These findings have great potential importance since they represent the first demonstration of a direct effect of androgens on the transcriptional process in the Sertoli cell. Furthermore, the results of these studies constitute further evidence that the Sertoli cell is a target for androgen action in the testis. ^

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Previously reported androgen receptor concentrations in rat testis and testicular cell types have varied widely. In the studies reported here a nuclear exchange assay was established in rat testis in which exchange after 86 hours at 4$\sp\circ$C was greater than 85% complete and receptor was stable. Receptor concentration per DNA measured by exchange declined between 15 and 25 days of age in the rat testis, then increased 4-fold during sexual maturation. Proliferation of germ cells which had low receptor concentration appeared to account for the early decline in testicular receptor concentration, whereas increase in receptor number per Sertoli cell between 25 and 35 days of age contributed to the later increase. Increase in Leydig cell number during maturation appeared to account for the remainder of the increase due to the high receptor concentration in these cells. Detailed studies showed that other possible explanations for changes in receptor number (e.g. shifts in receptor concentration between the cytosol and nuclear subcellular compartments or changes in the affinity of the receptor for its ligands) were not likely.^ Androgen receptor dynamics in testicular cells showed rapid, specific uptake of ($\sp3$H) -testosterone that was easily blocked by unlabeled testosterone (RA of 7 nM in both cell types), and medroxyprogesterone acetate (RA of 28 and 16 nM in Sertoli and peritubular cells, respectively), but not as well by the anti-androgens cyproterone acetate (RA of 116 and 68 nM) and hydroxyflutamide (RA of 300 and 180 nM). The affinity of the receptor for the ligand dimethylnortestosterone was similar in the two cell types (K$\rm\sb{d}$ values of 0.78 and 0.71 nM for Sertoli and peritubular cells) and was virtually identical with the affinity of the whole testis receptor (0.89 nM). Medroxyprogesterone acetate and testosterone significantly increased nuclear androgen receptor concentration relative to untreated controls in Sertoli and peritubular cells, whereas hydroxyflutamide and cyproterone acetate did not. Despite the different embryological origins of peritubular and Sertoli cells, their responses to both androgens and anti-androgens were similar. In addition, these studies suggest that peritubular cells are as likely as Sertoli cells to be primary androgen targets. ^

Relevância:

60.00% 60.00%

Publicador:

Resumo:

Pem, a member of the PEPP homeobox family, is expressed in somatic cells in male and female reproductive tissues. In the adult murine testis, Pem is specifically expressed in Sertoli cells, where it is restricted to stages IV–VIII of the seminiferous epithelial cycle. To identify Pem's function in Sertoli cells, transgenic mice were generated that express Pem in Sertoli cells during all stages of the seminiferous epithelial cycle. This resulted in an increase in double-strand DNA breaks in preleptotene spermatocytes and single-strand DNA breaks in elongating spermatids. My results suggest that Pem regulates Sertoli-cell genes that encode secreted or cell-surface proteins that serve to control premeiotic DNA replication, DNA repair, and/or chromatin remodeling in the adjacent germ cells. Three additional transgenic mouse containing varying lengths of the Pem male-specific promoter (Pp) were generated to identify the sequences responsible for regulating Pem expression in the testis and epididymis. My analysis suggests that there are at least two regulatory regions in the Pem Pp. In the testis, region II directs androgen-dependent expression specifically in Sertoli cells whereas region I fine-tunes stage-specific expression by acting as a negative regulator. In the epididymis, region II confers androgen-dependent, developmentally-regulated expression in the caput whereas region I prevents inappropriate expression in the corpus. I also report the identification and characterization of two human PEPP family members related to Pem that I have named hPEPP1 and hPEPP2. The hPEPP1 and hPEPP2 homeodomains are more closely related to PEPP subfamily homeodomains than to any other homeodomain subfamily. Both genes are localized to the specific region of the human X chromosome that shares synteny with the region on the murine X chromosome containing three PEPP homeobox genes, Pem, Psx-1, and Psx-2. hPEPP1 and hPEPP2 mRNA expression is restricted to the testis but is aberrantly expressed in tumor cells of different origins, analogous to the expression pattern of Pem but not of Psx-1 or Psx-2. Unlike all known PEPP members, neither hPEPP1 nor hPEPP2 are expressed in placenta, which suggests that the regulation of the PEPP family has undergone significant alteration since the split between hominids and rodents. ^

Relevância:

60.00% 60.00%

Publicador:

Resumo:

The X-linked mouse Rhox gene cluster contains over 30 homeobox genes that are candidates to regulate multiple steps in male and female gametogenesis. The founding member of the Rhox gene cluster, Rhox5, is an androgen-dependent gene expressed in Sertoli cells that promotes the survival and differentiation of the adjacent male germ cells. To decipher downstream signaling pathways of Rhox5, I used in vivo and in vitro microarray profiling to identify and characterize downstream targets of Rhox5 in the testis. This led to the identification of many Rhox5 -regulated genes, two of which I focused on in more detail. One of them, Unc5c, encodes a pro-apoptotic receptor with tumor suppressor activity that I found is negatively regulated by Rhox5 through a Rhox5-response element in the Unc5c 5' untranslated region (5' UTR). Examination of other mouse Rhox family members revealed that Rhox2 and Rhox3 also have the ability to downregulate Unc5c expression. The human RHOX protein RHOXF2 also had this ability, indicating that Unc5c repression is a conserved Rhox-dependent response. The repression of Unc5c expression by Rhox5 may, in part, mediate Rhox5's pro-survival function in the testis, as I found that Unc5c mutant mice have decreased germ cell apoptosis in the testis. This along with my other data leads me to propose a model in which Rhox5 is a negative regulator upstream of Unc5c in a Sertoli-cell pathway that promotes germ-cell survival. The other Rhox5-regulated gene that I studied in detail is insulin II (Ins2). Several lines of evidence, including electrophoretic mobility shift anaylsis, promoter mutagenesis, and chromatin immuoprecipitation analysis indicated that Ins2 is a direct target of Rhox5. Structure-function analysis identified homeodomain residues and the RHOX5 amino-terminal domain crucial for conferring Ins2 inducibility. Rhox5 regulates not only the Ins2 gene but also genes encoding other secreted proteins regulating metabolism (adiponectin and resistin), the rate-liming enzyme for monosaturated fatty acid biosynthesis (SCD-1), and transcription factors crucial for regulating metabolism (the nuclear hormone receptor PPARγ). I propose that the regulation of some or all of these molecules in Sertoli cells is responsible for the Rhox5-dependent survival of the adjacent germ cells. ^