Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Chemosphere ; 290: 133267, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34914963

ABSTRACT

The adverse outcome pathway (AOP) has been recently proposed as an effective framework for chemical risk assessment. The AOP framework offers the advantage of effectively integrating individual in vitro studies and in silico prediction models. Thus, the development of an effective testing method to measure key events caused by chemicals is essential for chemical risk assessment through a fully developed AOP framework. We developed a human cell-based estrogen receptor α (ERα) dimerization assay using the bioluminescence resonance energy transfer (BRET) technique and evaluated the ERα dimerization activities of 72 chemicals. Fifty-one chemicals were identified to mediate dimerization of ERα, and the BRET-based ERα dimerization assay could effectively measure the events that mediated dimerization of ERα by the estrogenic chemicals. These results were compared with the results of pre-existing assay to determine whether the BRET-based ERα dimerization assay could be employed as an in vitro test method to provide scientific information for explaining key events as a part of the AOP framework. Consequently, we propose that the BRET-based ERα dimerization assay is suitable for measuring the chemical-mediated dimerization of ERα, a key event in the AOP framework for cellular-level risk assessment of estrogenic chemicals.


Subject(s)
Adverse Outcome Pathways , Endocrine Disruptors , Dimerization , Endocrine Disruptors/toxicity , Energy Transfer , Estrogen Receptor alpha/metabolism , Humans
2.
Cell Metab ; 32(4): 591-604.e7, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32738205

ABSTRACT

Regulatory T cells (Tregs) are vital for the maintenance of immune homeostasis, while their dysfunction constitutes a cardinal feature of autoimmunity. Under steady-state conditions, mitochondrial metabolism is critical for Treg function; however, the metabolic adaptations of Tregs during autoimmunity are ill-defined. Herein, we report that elevated mitochondrial oxidative stress and a robust DNA damage response (DDR) associated with cell death occur in Tregs in individuals with autoimmunity. In an experimental autoimmune encephalitis (EAE) mouse model of autoimmunity, we found a Treg dysfunction recapitulating the features of autoimmune Tregs with a prominent mtROS signature. Scavenging of mtROS in Tregs of EAE mice reversed the DDR and prevented Treg death, while attenuating the Th1 and Th17 autoimmune responses. These findings highlight an unrecognized role of mitochondrial oxidative stress in defining Treg fate during autoimmunity, which may facilitate the design of novel immunotherapies for diseases with disturbed immune tolerance.


Subject(s)
Autoimmunity/immunology , Mitochondria/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
3.
Methods Mol Biol ; 1880: 331-340, 2019.
Article in English | MEDLINE | ID: mdl-30610708

ABSTRACT

Fluidic shear stress applied to epithelial cells inside the kidney tubules affects cell size in an autophagy-related manner. Here, we describe the technical equipment that we routinely use to apply shear stress on cells, as well as immunoblotting, immunofluorescence, and three-dimensional cell volume reconstruction techniques used in analysis of the influence of this stress on cells and cellular components. By pointing out details of experimental techniques and potential pitfalls, this review will serve as a guide for those interested in study of how shear stress influences cells.


Subject(s)
Autophagy/physiology , Biological Assay/methods , Cell Size , Epithelial Cells/cytology , Imaging, Three-Dimensional/methods , Animals , Biological Assay/instrumentation , Cell Line , Dogs , Epithelial Cells/physiology , Humans , Imaging, Three-Dimensional/instrumentation , Mice , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Shear Strength/physiology , Software , Stress, Mechanical
4.
Food Microbiol ; 67: 106-115, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28648287

ABSTRACT

The objective of this study was to evaluate the biodiversity of Aspergillus section Nigri populations from Cyprus vineyards by morphological, toxigenic and phylogenetic analysis. Aspergillus section Nigri populations were isolated from grapes of the varieties 'Maratheftiko' and 'Cabernet Sauvignon' originating from six growing regions of Cyprus during 2010 and 2011 years. The isolation frequency of Aspergillus section Nigri from grape samples was 43.3% and a total of 284 isolates were selected for further analyses based on the macroscopic characteristics of black aspergilli. The isolates were characterized by sequencing analysis of the calmodulin gene in order to identify species responsible for ochratoxin A (OTA) production. The phylogenetic analysis showed that the isolates were grouped in three major clusters. The A. tubingensis cluster included 262 isolates (92.25%), the A. niger cluster included 15 isolates identified as A. niger (5.3%) and 6 isolates identified as A. welwitschiae (2.1%). One isolate was classified as A. carbonarius (0.35%) and was grouped in a cluster together with the reference isolates of A. carbonarius, A. sclerotioniger, A. sclerotiocarbonarius and A. ibericus. All the isolates were evaluated for their ochratoxigenic ability by HPLC coupled with a fluorescence detector (HPLC-FLD) and the positive isolates were re-examined using ultra high-performance liquid chromatography with tandem mass spectrometry (UPLC-MS/MS). The Aspergillus carbonarius isolate produced an average quantity of 1436.1 ng OTA/g Czapek Yeast Agar (CYA); From the A. niger strains three isolates (20%) produced OTA and only one isolate from A. welwitschiae (16.7%) was proved ochratoxigenic with toxin production average at 23.9 ng/g and 9.1 ng/g CYA respectively. Grape must samples derived from the collected berries were also analyzed for OTA and none of the samples were found contaminated with the mycotoxin. The results showed that the geographic area and the meteorological conditions had no significant effect on the incidence and the distribution of black aspergilli in this 2-year project. However, absence of rainfall and low humidity during the harvesting period were critical for the low incidence of the ochratoxigenic A. carbonarius on grapes.


Subject(s)
Aspergillus/isolation & purification , Biodiversity , Ochratoxins/biosynthesis , Vitis/microbiology , Aspergillus/classification , Aspergillus/genetics , Aspergillus/metabolism , Cyprus , Food Contamination/analysis , Fruit/microbiology , Phylogeny , Wine/analysis
5.
Reproduction ; 152(6): 613-628, 2016 12.
Article in English | MEDLINE | ID: mdl-27601715

ABSTRACT

Follicle-stimulating hormone (FSH) stimulates the proliferation of immature Sertoli cells through the activation of PI3K/AKT/mTORC1 and MEK/ERK1/2 pathways. Mature Sertoli cells stop proliferating and respond to FSH by stimulating cAMP production. To gain insight into possible mechanisms involved in this switch as well as the impact of paracrine factors that stimulate cell proliferation, we analyzed the effects of FSH and relaxin on intracellular signaling pathways involved with proliferation and differentiation in Sertoli cells from 15-day-old rats, which are close to the transition between the two stages. FSH stimulated 3H-thymidine incorporation and cyclin D1 expression, changes associated with proliferation. In contrast, FSH inhibited AKT and ERK1/2 phosphorylation, activated cAMP production and induced changes in several cell cycle genes that were compatible with differentiation. Relaxin also stimulated 3H-thymidine incorporation but increased phosphorylation of ERK1/2 and AKT. When both hormones were added simultaneously, relaxin attenuated FSH-mediated inhibition of ERK1/2 and AKT phosphorylation and FSH-mediated activation of cAMP production. FSH but not relaxin increased CREB phosphorylation, and relaxin but not FSH shifted NF-κB expression from the cytoplasm to the nucleus. Relaxin did not inhibit the effects of FSH on inhibin α and Bcl2 expression. We propose that at this time of Sertoli cell development, FSH starts to direct cells to differentiation through activation of cAMP/CREB and inhibition of ERK1/2 and AKT pathways. Relaxin counteracts FSH signaling through the inhibition of cAMP and activation of ERK1/2, AKT and NF-κB, but does not block the differentiation process triggered by FSH.


Subject(s)
Cell Proliferation/drug effects , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Relaxin/pharmacology , Sertoli Cells/cytology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Hormones/pharmacology , Male , Phosphorylation/drug effects , Rats , Rats, Wistar , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Signal Transduction/drug effects
6.
Mol Cell Endocrinol ; 430: 12-24, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27107935

ABSTRACT

The aim of the present study was to characterize the mechanism underlying estrogen effects on the androgen-independent prostate cancer cell line PC-3. 17ß-estradiol and the ERß-selective agonist DPN, but not the ERα-selective agonist PPT, increased the incorporation of [methyl-(3)H]thymidine and the expression of Cyclin D2, suggesting that ERß mediates the proliferative effect of estrogen on PC-3 cells. In addition, upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by 17ß-estradiol and DPN were blocked by the ERß-selective antagonist PHTPP in PC-3 cells. Upregulation of Cyclin D2 and incorporation of [methyl-(3)H]thymidine induced by DPN were also blocked by PKF118-310, a compound that disrupts ß-catenin-TCF (T-cell-specific transcription factor) complex, suggesting the involvement of ß-catenin in the estradiol effects in PC-3 cells. A diffuse immunostaining for non-phosphorylated ß-catenin was detected in the cytoplasm of PC-3 cells. Low levels of non-phosphorylated ß-catenin immunostaining were also detected near the plasma membrane and in nuclei. Treatment of PC-3 cells with 17ß-estradiol or DPN markedly increased non-phosphorylated ß-catenin expression. These effects were blocked by pretreatment with the ERß-selective antagonist PHTPP, PI3K inhibitor Wortmannin or AKT inhibitor MK-2206, indicating that ERß-PI3K/AKT mediates non-phosphorylated ß-catenin expression. Cycloheximide blocked the DPN-induced upregulation of non-phosphorylated ß-catenin, suggesting de novo synthesis of this protein. In conclusion, these results suggest that estrogen may play a role in androgen-independent prostate cancer cell proliferation through a novel pathway, involving ERß-mediated activation of ß-catenin.


Subject(s)
Estrogen Receptor beta/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , beta Catenin/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin D2/metabolism , Cycloheximide/pharmacology , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Humans , Male , Nitriles/pharmacology , Phenols/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Thymidine/metabolism
7.
J Mol Endocrinol ; 54(3): 339-49, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26069273

ABSTRACT

Expression of the estrogen receptor ESR1 is higher in the corpus than it is in the initial segment/caput and cauda of the epididymis. ESR1 immunostaining in the corpus has been localized not only in the nuclei but also in the cytoplasm and apical membrane, which indicates that ESR1 plays a role in membrane-initiated signaling. The present study investigated whether ESR1 mediates the activation of rapid signaling pathways by estradiol (E2) in the epididymis. We investigated the effect of E2 and the ESR1-selective agonist (4,4',4''-(4-propyl-(1H)-pyrazole-1,3,5-triyl)trisphenol (PPT) on the activation of extracellular signal-regulated protein kinases (ERK1/2), CREB protein, and ETS oncogene-related protein (ELK1). Treatment with PPT did not affect ERK1/2 phosphorylation in the cauda, but it rapidly increased ERK1/2 phosphorylation in the initial segment/caput and corpus of the epididymis. PPT also activated CREB and ELK1 in the corpus of the epididymis. The PPT-induced phosphorylation of ERK1/2, CREB, and ELK1 was blocked by the ESR1-selective antagonist MPP and by pretreatment with a non-receptor tyrosine kinase SRC inhibitor, an EGFR kinase inhibitor, an MEK1/2 inhibitor, and a phosphatidylinositol-3-kinase inhibitor. In conclusion, these results indicate that the corpus, which is a region with high expression of the estrogen receptor ESR1, is a major target in the epididymis for the activation of rapid signaling by E2. The sequence of events that follow E2 interaction with ESR1 includes the SRC-mediated transactivation of EGFR and the phosphorylation of ERK1/2, CREB, and ELK1. This rapid estrogen signaling may modulate gene expression in the corpus of the epididymis, and it may play a role in the dynamic microenvironment of the epididymal lumen.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Epididymis/enzymology , Estrogen Receptor alpha/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , ets-Domain Protein Elk-1/metabolism , Animals , Estradiol/physiology , Estrogen Receptor alpha/agonists , MAP Kinase Signaling System , Male , Phenols/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Pyrazoles/pharmacology , Rats, Wistar
8.
Spermatogenesis ; 4: e28138, 2014.
Article in English | MEDLINE | ID: mdl-25225624

ABSTRACT

The identification of the hormones and other factors regulating Sertoli cell survival, proliferation, and maturation in neonatal, peripubertal, and pubertal life remains one of the most critical questions in testicular biology. The regulation of Sertoli cell proliferation and differentiation is thought to be controlled by cell-cell junctions and a set of circulating and local hormones and growth factors. In this review, we will focus on receptors and intracellular signaling pathways activated by androgen, follicle-stimulating hormone, thyroid hormone, activin, retinoids, insulin, insulin-like growth factor, relaxin, and estrogen, with special emphasis on estrogen receptors. Estrogen receptors activate intracellular signaling pathways that converge on cell cycle and transcription factors and play a role in the regulation of Sertoli cell proliferation and differentiation.

9.
Biomed Res Int ; 2014: 487129, 2014.
Article in English | MEDLINE | ID: mdl-24696852

ABSTRACT

Exercise training reduces sympathetic activity in hypertensive humans and rats. We hypothesized that the swimming exercise would change the neurotransmission in the rostral ventrolateral medulla (RVLM), a key region involved in sympathetic outflow, and hemodynamic control in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats. Bilateral injections of kynurenic acid (KYN) were carried out in the RVLM in sedentary- (S-) or exercised- (E-) SHR and WKY rats submitted to swimming for 6 weeks. Rats were α-chloralose anesthetized and artificially ventilated, with Doppler flow probes around the lower abdominal aorta and superior mesenteric artery. Injections into the RVLM were made before and after i.v. L-NAME (nitric oxide synthase, NOS, inhibitor). Injections of KYN into the RVLM elicited a major vasodilation in the hindlimb more than in the mesenteric artery in E-SHR compared to S-SHR, but similar decrease in arterial pressure was observed in both groups. Injections of KYN into the RVLM after i.v. L-NAME attenuated the hindlimb vasodilation evoked by KYN and increased the mesenteric vasodilation in E-SHR. Swimming exercise can enhance the hindlimb vasodilation mediated by peripheral NO release, reducing the activation of neurons with EAA receptors in the RVLM in SHR.


Subject(s)
Hemodynamics , Medulla Oblongata/physiology , Physical Conditioning, Animal , Receptors, Glutamate/metabolism , Swimming , Animals , Blood Pressure/drug effects , Cardiovascular System/drug effects , Glutamic Acid/pharmacology , Heart Rate/drug effects , Hemodynamics/drug effects , Kynurenic Acid/pharmacology , Medulla Oblongata/drug effects , Nitric Oxide/metabolism , Rats, Inbred SHR , Rats, Inbred WKY
10.
Mol Cell Endocrinol ; 382(1): 84-96, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24056172

ABSTRACT

The aim of the present study was to investigate the role of each estrogen receptors on the regulation of proteins involved with proliferation and differentiation of Sertoli cells from 15-day-old rats. Activation of ESR1 by 17ß-estradiol (E2) and ESR1-selective agonist PPT increased CCND1 expression, and this effect was dependent on NF-kB activation. E2 and the ESR2-selective agonist DPN, but not PPT, increased, in a PI3K and CREB-dependent manner, the expression of CDKN1B and the transcription factors GATA-1 and DMRT1. Analyzing the expression of ESR1 and ESR2 in different stages of development of Sertoli cells, we observed that the ESR1/ESR2 ratio decreased with age, and this ratio seems to be important to determine the end of cell proliferation and the start of cell differentiation. In Sertoli cells from 15-day-old rats, the ESR1/ESR2 ratio favors the effect of ESR1 and the activation of this receptor increased [Methyl-(3)H]thymidine incorporation. We propose that in Sertoli cells from 15-day-old rats E2 modulates Sertoli cell proliferation through ESR1/NF-kB-mediated increase of CCND1, and cell cycle exit and differentiation through ESR2/CREB-mediated increase of CDKN1B, GATA-1 and DMRT1. The present study reinforces the important role of estrogen for normal testis development.


Subject(s)
Cell Differentiation , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Estradiol/pharmacology , GATA1 Transcription Factor/metabolism , I-kappa B Proteins/metabolism , Male , Models, Biological , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nitriles/pharmacology , Phenols/pharmacology , Phosphorylation/drug effects , Protein Transport/drug effects , Pyrazoles/pharmacology , Rats , Rats, Wistar , Sertoli Cells/drug effects , Thymidine/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects
11.
Spermatogenesis ; 3(1): e23181, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23687614

ABSTRACT

The Wnt/ß-catenin signaling pathway controls several biological processes throughout development and adult life. Dysregulation of Wnt/ß-catenin signaling underlies a wide range of pathologies in animals and humans, including cancer in different tissues. In this review, we provide an update of the Wnt/ß-catenin signaling pathway and the possible roles of the Wnt/ß-catenin signaling in the biology of testis, epididymis and prostate. Data from our laboratory suggest the involvement of 17ß-estradiol and estrogen receptors (ERs) on the regulation of ß-catenin expression in rat Sertoli cells. We also provide emerging evidences of the involvement of Wnt/ß-catenin pathway in testis and prostate cancer. Our understanding of the role of Wnt/ß-Catenin signaling in male reproductive tissues is still evolving, and several questions are open to be addressed in the future.

12.
Ital J Anat Embryol ; 118(1 Suppl): 26-8, 2013.
Article in English | MEDLINE | ID: mdl-24640565

ABSTRACT

Immature Sertoli cells proliferate and several factors affect their number, including the follicle stimulating hormone (FSH), testosterone, estradiol and several paracrine growth factors. Using a primary culture of Sertoli cells isolated from 15-day old Wistar rats we have shown that relaxin stimulates Sertoli cell proliferation through the activation of MEK/ERK1/2 and PI3K/AKT pathways. In contrast, FSH inhibited both ERK1/2 and AKT phosphorylation. Furthermore, FSH strongly increased cAMP production, whereas relaxin inhibited basal cAMP production. Our results indicate that in rat Sertoli cells from 15-day old rats relaxin and FSH affect the same signaling pathways in opposite directions. Interplay between both hormones may be important to control the proliferation and differentiation of Sertoli cells.


Subject(s)
MAP Kinase Signaling System/physiology , Relaxin/physiology , Sertoli Cells/cytology , Sertoli Cells/physiology , Animals , Cell Proliferation , Follicle Stimulating Hormone/physiology , Male , Primary Cell Culture , Rats , Rats, Wistar
13.
Ital J Anat Embryol ; 118(1 Suppl): 29-31, 2013.
Article in English | MEDLINE | ID: mdl-24640566

ABSTRACT

Spermatogenesis is controlled by FSH, testosterone and paracrine factors produced by Sertoli cells. The knockout of relaxin decreases sperm maturation in mice. Studies from our laboratory have shown that relaxin and its receptor RXFP1 are expressed in rat Sertoli cells, and exogenous relaxin stimulates Sertoli cell proliferation. Relaxin receptors are also detected in the rat germ cells at specific stages of development. Relaxin could therefore affect spermatogenesis either indirectly, by stimulating Sertoli cell proliferation, or directly, by affecting germ cells. The aim of the present study was to explore a role of relaxin at specific stages of spermatogenesis using a co-culture of rat Sertoli and germ cells. Relaxin seems to increase the number of pre-meiotic and meiotic cells.


Subject(s)
Relaxin/physiology , Sertoli Cells/cytology , Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatozoa/cytology , Spermatozoa/physiology , Animals , Cells, Cultured , Coculture Techniques , Male , Rats
14.
Eur J Pharmacol ; 691(1-3): 283-91, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22819701

ABSTRACT

Regulation of Sertoli cell number is a key event to determine normal spermatogenesis. We have previously shown that relaxin and its G-protein coupled receptor RXFP1 are expressed in rat Sertoli cells, and that relaxin stimulates Sertoli cell proliferation. This study examined the mechanisms underlying the mitogenic effect of relaxin in a primary culture of Sertoli cells removed from testes of immature rats. Stimulation with exogenous relaxin increased Sertoli cell number and the expression of the proliferating cell nuclear antigen (PCNA), but did not affect the mRNA level of the differentiation markers cadherins 1 and 2. Relaxin-induced Sertoli cell proliferation was blocked by inhibition of MEK/ERK1/2 or PI3K/AKT pathways, but not by inhibition of PKC or EGFR activity. Relaxin induced a rapid and transient activation of ERK1/2 phosphorylation, which was MEK and SRC-dependent, and involved upstream activation of G(i). AKT activation could be detected 5 min after relaxin stimulation, and was still detected after 24h of stimulation with relaxin. Relaxin-induced AKT phosphorylation was G(i)- but not PKA-dependent, and it was blocked by both PI3K and MEK inhibitors. In conclusion, the mitogenic effect of relaxin in Sertoli cell involves coupling to G(i) and activation of both MEK/ERK1/2 and PI3K/AKT pathways.


Subject(s)
Intracellular Space/drug effects , Intracellular Space/metabolism , Relaxin/pharmacology , Sertoli Cells/cytology , Sertoli Cells/drug effects , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Sertoli Cells/metabolism
15.
Biol Reprod ; 86(4): 108, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22219213

ABSTRACT

The aim of the present study was to investigate the intracellular signaling events downstream of the classical estrogen receptors (ESRs) and G protein-coupled estrogen receptor 1 (GPER) involved in regulation of proliferation and apoptosis of rat Sertoli cells, in which we have previously described ESR1, ESR2, and GPER. ESRs play a role in Sertoli cell proliferation, and GPER, but not ESRs, plays a role modulating gene expression involved with apoptosis. The present study shows that 17beta-estradiol (E2) and the GPER-selective agonist G-1 rapidly activate phosphatidylinositol 3-kinase (PIK3)/serine threonine protein kinase (AKT) and cyclic AMP response element-binding (CREB) phosphorylation. E2 and the ESR1-selective agonist 4,4',4″-(4-propyl-(1H)-pyrazole-1,3,5-triyl)trisphenol (PPT) increase the expression of cyclin D1 (CCND1), whereas the ESR2-selective agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN) and G-1 do not change the expression of this protein, suggesting that ESR1 is the upstream receptor regulating Sertoli cell proliferation. E2- or PPT-ESR1, through activation of epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase 3/1 (MAPK3/1) and PIK3 pathways, induces upregulation of CCND1. KG-501, the compound that disrupts the phospho-CREB/CREB binding protein (CBP) complex, does not change E2- or PPT-ESR1-mediated CCND1 expression, suggesting that phospho-CREB/cyclic AMP response element/CBP is not involved in the expression of this protein. E2- or G-1-GPER, through activation of EGFR/MAPK3/1 and PIK3 pathways, may be involved in the upregulation of antiapoptotic proteins BCL2 and BCL2L2. E2- or G-1-GPER/EGFR/MAPK3/1/phospho-CREB decreases BAX expression. Taken together, these results show a differential effect of E2-GPER on the CREB-mediated transcription of proapoptotic and antiapoptotic genes of the same BCL2 gene family. ESR1 and GPER can mediate the rapid E2 actions in the Sertoli cells, which in turn can modulate nuclear transcriptional events important for Sertoli cell function and maintenance of normal testis development and homeostasis. Our findings are important to clarify the role of estrogen in a critical period of testicular development, and to direct further studies, which may contribute to better understanding of the causes of male infertility.


Subject(s)
Apoptosis/physiology , Estradiol/physiology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Sertoli Cells/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Phosphatidylinositol 3-Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Receptors, Estrogen/agonists , Receptors, G-Protein-Coupled/agonists , Signal Transduction/physiology , Up-Regulation
16.
J Androl ; 32(6): 600-13, 2011.
Article in English | MEDLINE | ID: mdl-21441425

ABSTRACT

Estrogens play key roles in the development and maintenance of male reproductive function and fertility. In this review, we briefly describe the localization and function of estrogen receptors ESR1 and ESR2 (also known as ERα and ERß, respectively) and the expression of G protein-coupled estrogen receptor-1 (GPER, formerly known as GPR30) in efferent ductules and epididymis. The efferent ductules present the highest levels of ESR1 and ESR2 in the male reproductive system, and represent a major target of estrogen action. In efferent ductules, ESR1 has a crucial role in the regulation of fluid reabsorption, and in the epididymis the receptor helps to maintain fluid osmolality and pH. ESR1 expression in the epididymal epithelium shows considerable variation among species, but differences in laboratory techniques may also contribute to this variation. Here we report that Esr1 mRNA and protein are higher in corpus than in other regions of the rat epididymis. The mRNA level for Gper was also higher in corpus. Although ESR1 is expressed constitutively in efferent ductules and down-regulated by estrogen, in the epididymis, both testosterone (T) and estradiol (E2) may regulate its expression. T and E2 are, respectively, higher and lower in the corpus than in the initial segment/caput and cauda regions. It is important to determine the expression of GPER, ESR1, androgen receptor, and their respective cofactors in specific cell types of this tissue, as well as the intracellular signaling pathways involved in efferent ductules and epididymis. These studies will help to explain the consequences of exposures to environmental endocrine disruptors and provide potential targets for the development of a male contraceptive.


Subject(s)
Ejaculatory Ducts/metabolism , Epididymis/metabolism , Estrogens/metabolism , Receptors, Estrogen/metabolism , Androgens/metabolism , Animals , Cats , Cattle , Cricetinae , Dogs , Ejaculatory Ducts/cytology , Epididymis/cytology , Estrogens/analysis , Haplorhini , Humans , Male , Mice , Rats , Receptors, G-Protein-Coupled/metabolism , Swine
17.
Biol Reprod ; 84(1): 52-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20826728

ABSTRACT

Estrogen plays a key role in maintaining the morphology and function of the efferent ductules. We previously demonstrated that the antiestrogen fulvestrant markedly affected gene expression in the rat efferent ductules. The mechanism of fulvestrant action to modulate gene expression may involve not only the blockade of ESR1 and ESR2 estrogen receptors, but also the activation of ESR1 and ESR2 when the receptors are tethered to AP-1 or SP1 transcription factors, or the activation of the G protein-coupled estrogen receptor 1. We therefore compared the effects of two strategies to interfere with estrogen action in the rat efferent ductules: treatment with fulvestrant or with the aromatase inhibitor anastrozole. Whereas fulvestrant markedly increased Mmp7 and Spp1, and reduced Nptx1 mRNA levels, no changes were observed with anastrozole. Fulvestrant caused changes in epithelial morphology that were not seen with anastrozole. Fulvestrant shifted MMP7 immunolocalization in the epithelial cells from the supranuclear to the apical region; this effect was less pronounced with anastrozole. In vitro studies of (35)S-methionine incorporation showed that protein release was increased, whereas tissue protein content in the efferent ductules of fulvestrant-treated rats was decreased. Although fulvestrant markedly affected gene expression, no changes were observed on AP-1 and SP1 DNA-binding activity. The blockade of ESRs seems to be the major reason explaining the differences between both treatments. At least some of the effects of fulvestrant appear to result from compensatory mechanisms activated by the dramatic changes caused by ESR1 blockade.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Ejaculatory Ducts/drug effects , Estradiol/analogs & derivatives , Gene Expression Regulation/drug effects , Nitriles/pharmacology , Triazoles/pharmacology , Anastrozole , Animals , Ejaculatory Ducts/metabolism , Estradiol/blood , Estradiol/pharmacology , Fulvestrant , Male , Rats , Rats, Wistar , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Testosterone/blood , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
18.
Spermatogenesis ; 1(4): 318-324, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22332115

ABSTRACT

In this review, we will present an overview of estrogen actions in the testis from immature and adult animals, with special emphasis on signaling mechanisms involved in the 17ß-estradiol regulation of Sertoli cell function in immature rats. 17ß-estradiol activates Sertoli cell proliferation in immature rats by a mechanism that involves the translocation of the estrogen receptors ESR1 and ESR2 to the plasma membrane, phosphorylation of epidermal growth factor receptor and activation of mitogen-activated protein kinase 3/1. Activation of the G protein-coupled estrogen receptor (GPER) also induces phosphorylation of mitogen-activated protein kinase 3/1 via epidermal growth factor receptor transactivation, which in turn increases expression of the antiapoptotic protein BCL2 and decreases the expression of proapoptotic protein BAX, indicating an antiapoptotic role of E2-GPER in immature rat Sertoli cells. In conclusion, ESRs and GPER can mediate rapid 17ß-estradiol signaling in Sertoli cells, and modulate transcriptional events important for Sertoli cell function and maintenance of normal testis development and homeostasis. Our findings are important to clarify the role of estrogen in a critical period of testicular development and to direct further studies, which may contribute to better understand the causes of male infertility.

19.
Biol Reprod ; 83(2): 307-17, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20445128

ABSTRACT

The aim of the present study was to investigate the expression and signaling of the G protein-coupled estrogen receptor 1 (GPER) in cultured immature rat Sertoli cells--in which we have previously described the classical estrogen receptors (ESR1 and ESR2). Expression of GPER in cultured Sertoli cells from 15-day-old rats was detected by RT-PCR and immunoassays. Gper transcripts also were present in testes from 5-, 15-, and 120-day-old rats. Short-term treatment of Sertoli cells with 17beta-estradiol (E2), the GPER agonist G-1, or the ESR antagonist ICI 182,780 (ICI) rapidly activated MAPK3/1 (ERK1/2), even after down-regulation of ESR1 and ESR2, suggesting a role for GPER in the rapid E2 action in these cells. MAPK3/1 phosphorylation induced by ICI or G-1 was blocked by pertussis toxin, selective inhibitor of the SRC family of protein tyrosine kinases, metalloprotease inhibitor, MAP2K1/2 inhibitor, and epidermal growth factor receptor (EGFR) kinase inhibitor. Furthermore, E2, but not G-1, induced up-regulation of cyclin D1 in the Sertoli cells. This effect was blocked by ICI. E2 and G-1 decreased BAX and increased BCL2 expression and these effects were blocked by MAP2K1/2 inhibitor and EGFR kinase inhibitor. The pretreatment with ICI did not block the effect of E2. Taken together, these results indicate that in Sertoli cells 1) GPER-mediated MAPK3/1 activation occurs via EGFR transactivation through G protein beta gamma subunits that promote SRC-mediated metalloprotease-dependent release of EGFR ligands, which bind to EGFR and lead to MAPK3/1 phosphorylation; 2) E2-ESRs play a role in Sertoli cell proliferation; and 3) E2-GPER may regulate gene expression involved with apoptosis. ESR and GPER may mediate actions important for Sertoli cell function and maintenance of normal testis development and homeostasis.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Sertoli Cells/physiology , Signal Transduction , Animals , Apoptosis/genetics , Cells, Cultured , Cyclopentanes , Enzyme Activation/drug effects , ErbB Receptors/physiology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Fulvestrant , Gene Expression Regulation/physiology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Quinolines , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/chemistry , Testis/chemistry , Testis/physiology
20.
Reproduction ; 139(1): 185-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812235

ABSTRACT

We have previously shown that the rat testis and vas deferens contain high levels of the relaxin receptor, RXFP1. The present study was undertaken to determine the expression of relaxin in these tissues, and the effect of exogenous relaxin on Sertoli cell proliferation and on the mRNA levels of some proteins that may contribute to epithelial secretion and tissue reorganization in the vas deferens. Relaxin mRNA levels in testis and vas deferens were much lower than in the prostate. Sertoli cells seem to be an important source of relaxin mRNA in testis. Relaxin immunoreactivity was detected in the seminiferous epithelium but not in the interstitial compartment. The relaxin precursor was expressed in the vas deferens, and relaxin immunoreactivity was detected in apical cells of the vas deferens. Castration, but not treatment with the anti-estrogen ICI 182,780, dramatically reduced relaxin mRNA levels in the prostate and vas deferens, and this effect was prevented by testosterone. Rxfp1 mRNA levels in the vas deferens and prostate were not affected by castration or treatment with ICI 182,780. Exogenous relaxin increased the incorporation of (3)H-thymidine in cultured Sertoli cells, and treatment of the vas deferens with 100 ng/ml relaxin increased the mRNA levels for the cystic fibrosis chloride channel (cystic fibrosis transmembrane regulator) about three times, and doubled mRNA levels for the inducible form of nitric oxide synthase and metalloproteinase 7. These results suggest that locally produced relaxin acts as an autocrine or paracrine agent in the testis and vas deferens to affect spermatogenesis and seminal fluid composition.


Subject(s)
Relaxin/metabolism , Testis/metabolism , Vas Deferens/metabolism , Aging , Animals , Cell Proliferation , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Estrogen Antagonists/pharmacology , Female , In Vitro Techniques , Male , Matrix Metalloproteinase 7/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Orchiectomy , Organ Specificity , Ovary/cytology , Ovary/metabolism , Pregnancy , Prostate/cytology , Prostate/drug effects , Prostate/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Relaxin/genetics , Sertoli Cells/metabolism , Testis/cytology , Testosterone/pharmacology , Vas Deferens/cytology , Vas Deferens/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...