Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Food Chem Toxicol ; 158: 112649, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34728246

ABSTRACT

Phthalates are synthetic chemicals widely used to make polyvinylchloride (PVC) soft and flexible. Of these, Di-(2-ethylhexyl) phthalate (DEHP) is the most commonly used, with high human exposure occurring as early as the fetal developmental stage and affecting the endocrine system. We focused on the perinatal DEHP effects on pituitary estrogen receptor (ER) expression in male rats, explored their impact on lactotroph and somatotroph cell growth, and evaluated the direct effects of this phthalate on pituitary cell cultures. Our results showed that DEHP perinatal exposure was unable to modify the ERα+ pituitary cell number from prepuberal rats, but increased ERß+ cells. In adulthood, the pituitary ERα+ cells underwent a slight decrease with ERß showing the greatest changes, and with a significant increase observed in somatotroph cells. Also, in vitro, DEHP reduced the ERα+ cells, increased the percentage of ERß+ pituitary cells and modified the Ki67 index, as well as decreasing the lactotrophs and increasing the somatotroph cells. In conclusion, the present study showed that DEHP induced ER expression changes in normal pituitary glands from male rats in in vivo and in vitro conditions, suggesting that DEHP could differentially modulate lactotroph and somatotroph cell growth, possibly as a consequence of ER imbalance.


Subject(s)
Diethylhexyl Phthalate/toxicity , Endocrine Disruptors/toxicity , Pituitary Gland , Prenatal Exposure Delayed Effects , Receptors, Estrogen/metabolism , Animals , Cell Proliferation/drug effects , Female , Lactotrophs/drug effects , Lactotrophs/metabolism , Male , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar , Somatotrophs/drug effects , Somatotrophs/metabolism
2.
Chemosphere ; 258: 127304, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32559490

ABSTRACT

Humans are exposed to numerous endocrine disruptors on a daily basis, which may interfere with endogenous estrogens, with Di-(2-ethylhexyl) phthalate (DEHP) being one of the most employed. The anterior pituitary gland is a target of 17ß-estradiol (E2) through the specific estrogen receptors (ERs) α and ß, whose expression levels fluctuate in the gland under different contexts, and the ERα/ß index is responsible for the final E2 effect. The aim of the present study was to evaluate in vivo and in vitro the DEHP effects on ERα and ß expression in the pituitary cell population, and also its impact on lactotroph and somatotroph cell growth. Our results revealed that perinatal exposure to DEHP altered the ERα and ß expression pattern in pituitary glands from prepubertal and adult female rats and increased the percentage of lactotroph cells in adulthood. In the in vitro system, DEHP down-regulated ERα and ß expression, and as a result increased the ERα/ß ratio and decreased the percentages of lactotrophs and somatotrophs expressing ERα and ß. In addition, DEHP increased the S + G2M phases, Ki67 index and cyclin D1 in vitro, leading to a rise in the lactotroph and somatotroph cell populations. These results showed that DEHP modified the pituitary ERα and ß expression in lactotrophs and somatotrophs from female rats and had an impact on the pituitary cell growth. These changes in ER expression may be a mechanism underlying DEHP exposure in the pituitary gland, leading to cell growth deregulation.


Subject(s)
Diethylhexyl Phthalate/toxicity , Phthalic Acids/toxicity , Receptors, Estrogen/metabolism , Animals , Cell Proliferation/drug effects , Diethylhexyl Phthalate/metabolism , Endocrine Disruptors/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Female , Lactotrophs/drug effects , Lactotrophs/metabolism , Pituitary Gland/drug effects , Rats
3.
J Endocrinol ; 246(1): 29-39, 2020 07.
Article in English | MEDLINE | ID: mdl-32302971

ABSTRACT

Serum prolactin levels gradually increase from birth to puberty in both male and female rats, with higher levels observed in female since the first days of life. The increase in lactotroph secretion was attributed to the maturation of prolactin-inhibiting and prolactin-releasing factors; however, those mechanisms could not fully explain the gender differences observed. Prolactin secretion from isolated lactotrophs, in the absence of hypothalamic control, also increases during the first weeks of life, suggesting the involvement of intra-pituitary factors. We postulate that pituitary transforming growth factor beta 1 (TGFß1) is involved in the regulation of prolactin secretion as well as in the gender differences observed at early postnatal age. Several components of the local TGFß1 system were evaluated during postnatal development (11, 23, and 45 days) in female and male Sprague-Dawley rats. In vivo assays were performed to study local TGFß1 activation and its impact on prolactin secretion. At day 11, female pituitaries present high levels of active TGFß1, concomitant with the highest expression of TGFß1 target genes and the phospho-Smad3 immunostaining in lactotrophs. The steady increase in prolactin secretion inversely correlates with active TGFß1 levels only in females. Dopamine and estradiol induce TGFß1 activation at day 11, in both genders, but its activation induces the inhibition of prolactin secretion only in females. Our findings demonstrate that: (1) TGFß1 activation is regulated by dopamine and estradiol; (2) the inhibitory regulation of local TGFß1 on prolactin secretion is gender specific; and (3) this mechanism is responsible, at least partially, for the gender differences observed being relevant during postnatal development.


Subject(s)
Transforming Growth Factor beta1/metabolism , Animals , Dopamine/pharmacology , Estradiol/pharmacology , Female , Lactotrophs/drug effects , Lactotrophs/metabolism , Male , Prolactin/metabolism , Rats , Rats, Sprague-Dawley , Sex Characteristics , Smad3 Protein/metabolism
4.
Neuroendocrinology ; 108(2): 84-97, 2019.
Article in English | MEDLINE | ID: mdl-30376668

ABSTRACT

Prolactinomas are increasingly viewed as a "problem of signal transduction." Consequently, the identification of factors and signaling pathways that control lactotrope cell turnover is needed in order to encourage new therapeutic developments. We have previously shown that prolactin (PRL) acts as a proapoptotic and antiproliferative factor on lactotropes, maintaining anterior pituitary cell homeostasis, which contrasts with the classical antiapoptotic and/or proliferative actions exerted by PRL in most other target tissues. We aimed to investigate the PRLR-triggered signaling pathways mediating these nonclassical effects of PRL in the pituitary. Our results suggest that (i) the PRLR/Jak2/STAT5 pathway is constitutively active in GH3 cells and contributes to PRL-induced apoptosis by increasing the Bax/Bcl-2 ratio, (ii) PRL inhibits ERK1/2 and Akt phosphorylation, thereby contributing to its proapoptotic effect, and (iii) the PI3K/Akt pathway participates in the PRL-mediated control of lactotrope proliferation. We hypothesize that the alteration of PRL actions in lactotrope homeostasis due to the dysregulation of any of the mechanisms of actions described above may contribute to the pathogenesis of prolactinomas.


Subject(s)
Apoptosis/drug effects , Janus Kinase 2/metabolism , Lactotrophs/metabolism , Prolactin/pharmacology , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Female , Lactotrophs/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Prolactin/antagonists & inhibitors , Receptors, Prolactin/metabolism , Signal Transduction/drug effects
5.
J Endocrinol ; 240(2): 99-110, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30400046

ABSTRACT

Ovarian steroids control a variety of physiological functions. They exert actions through classical nuclear steroid receptors, but rapid non-genomic actions through specific membrane steroid receptors have been also described. In this study, we demonstrate that the G-protein-coupled estrogen receptor (GPER) is expressed in the rat pituitary gland and, at a high level, in the lactotroph population. Our results revealed that ~40% of the anterior pituitary cells are GPER positive and ~35% of the lactotrophs are GPER positive. By immunocytochemical and immuno-electron-microscopy studies, we demonstrated that GPER is localized in the plasmatic membrane but is also associated to the endoplasmic reticulum in rat lactotrophs. Moreover, we found that local Gper expression is regulated negatively by 17ß-estradiol (E2) and progesterone (P4) and fluctuates during the estrus cycle, being minimal in proestrus. Interestingly, lack of ovarian steroids after an ovariectomy (OVX) significantly increased pituitary GPER expression specifically in the three morphologically different subtypes of lactotrophs. We found a rapid estradiol stimulatory effect on PRL secretion mediated by GPER, both in vitro and ex vivo, using a GPER agonist G1, and this effect was prevented by the GPER antagonist G36, demonstrating a novel role for this receptor. Then, the increased pituitary GPER expression after OVX could lead to alterations in the pituitary function as all three lactotroph subtypes are target of GPER ligand and could be involved in the PRL secretion mediated by GPER. Therefore, it should be taken into consideration in the response of the gland to an eventual hormone replacement therapy.


Subject(s)
Estradiol/pharmacology , Lactotrophs/metabolism , Pituitary Gland, Anterior/metabolism , Progesterone/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Estrogens/pharmacology , Female , Gene Expression/drug effects , Lactotrophs/drug effects , Lactotrophs/ultrastructure , Ovariectomy , Pituitary Gland, Anterior/cytology , Proestrus , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics
6.
J Cell Physiol ; 233(2): 1402-1413, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28542730

ABSTRACT

In this study, we focused on ERß regulation in the adenohypophysis under different estrogenic milieu, by analyzing whether ER modulates the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression and its subcellular localization on anterior pituitary glands from Wistar rats and GH3 lactosomatotroph cells that over-expressed ERß. ERß was regulated in a cyclic manner, and underwent dynamic changes throughout the estrous cycle, with decreased ERß+ cells in estrus and under E2 treatment, but increased in ovariectomized rats. In addition, the ERα/ß ratio increased in estrus and under E2 stimulation, but decreased in ovariectomized rats. Double immunofluorescence revealed that lactotroph and somatotroph ERß+ were significantly decreased in estrus. Also, variations in the PTEN expression was observed, which was diminished with high E2 conditions but augmented with low E2 milieu. The subcellular localization of this phosphatase was cell cycle-dependent, with remarkable changes in the immunostaining pattern: nuclear in arrested pituitary cells but cytoplasmic in stimulated cells, and responding differently to ER agonists, with only DPN being able to increase PTEN expression and retaining it in the nucleus. Finally, ERß over-expression increased PTEN with a noticeable subcellular redistribution, and with a significant nuclear signal increase in correlation with an increase of cells in G0/G1 phase. These results showed that E2 is able to inhibit ERß expression and suggests that the tumoral suppressor PTEN might be one of the signaling proteins by which E2, through ERß, acts to modulate pituitary cell proliferation, thereby adapting endocrine populations in relation with hormonal necessities.


Subject(s)
Cell Proliferation , Estrogen Receptor beta/metabolism , Estrous Cycle/metabolism , Lactotrophs/enzymology , PTEN Phosphohydrolase/metabolism , Somatotrophs/enzymology , Animals , Cells, Cultured , Estradiol/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogen Replacement Therapy , Female , G1 Phase , Lactotrophs/drug effects , Male , Nitriles/pharmacology , Ovariectomy , Rats, Wistar , Signal Transduction , Somatotrophs/drug effects , Transfection
7.
PLoS One ; 9(5): e97383, 2014.
Article in English | MEDLINE | ID: mdl-24859278

ABSTRACT

Anterior pituitary cell turnover occurring during female sexual cycle is a poorly understood process that involves complex regulation of cell proliferation and apoptosis by multiple hormones. In rats, the prolactin (PRL) surge that occurs at proestrus coincides with the highest apoptotic rate. Since anterior pituitary cells express the prolactin receptor (PRLR), we aimed to address the actual role of PRL in the regulation of pituitary cell turnover in cycling females. We showed that acute hyperprolactinemia induced in ovariectomized rats using PRL injection or dopamine antagonist treatment rapidly increased apoptosis and decreased proliferation specifically of PRL producing cells (lactotropes), suggesting a direct regulation of these cell responses by PRL. To demonstrate that apoptosis naturally occurring at proestrus was regulated by transient elevation of endogenous PRL levels, we used PRLR-deficient female mice (PRLRKO) in which PRL signaling is totally abolished. According to our hypothesis, no increase in lactotrope apoptotic rate was observed at proestrus, which likely contributes to pituitary tumorigenesis observed in these animals. To decipher the molecular mechanisms underlying PRL effects, we explored the isoform-specific pattern of PRLR expression in cycling wild type females. This analysis revealed dramatic changes of long versus short PRLR ratio during the estrous cycle, which is particularly relevant since these isoforms exhibit distinct signaling properties. This pattern was markedly altered in a model of chronic PRLR signaling blockade involving transgenic mice expressing a pure PRLR antagonist (TGΔ1-9-G129R-hPRL), providing evidence that PRL regulates the expression of its own receptor in an isoform-specific manner. Taken together, these results demonstrate that i) the PRL surge occurring during proestrus is a major proapoptotic signal for lactotropes, and ii) partial or total deficiencies in PRLR signaling in the anterior pituitary may result in pituitary hyperplasia and eventual prolactinoma development, as observed in TGΔ1-9-G129R-hPRL and PRLRKO mice, respectively.


Subject(s)
Apoptosis/drug effects , Lactotrophs/cytology , Lactotrophs/drug effects , Prolactin/pharmacology , Animals , Cell Proliferation/drug effects , Down-Regulation/drug effects , Estrous Cycle/drug effects , Female , Gene Knockout Techniques , Lactotrophs/metabolism , Mice , Prolactin/metabolism , Rats , Receptors, Prolactin/deficiency , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Signal Transduction/drug effects
8.
PLoS One ; 8(11): e81101, 2013.
Article in English | MEDLINE | ID: mdl-24236210

ABSTRACT

Cadmium (Cd) is a heavy metal of considerable occupational and environmental concern affecting wildlife and human health. Recent studies indicate that Cd, like other heavy metals, can mimic effects of 17ß-estradiol (E2) involving E2 receptor (ER) activation. Lactotrophs, the most abundant cell type in anterior pituitary gland, are the main target of E2, which stimulates cell proliferation and increases prolactin secretion through ERα. The aim of this work was to examine whether Cd at nanomolar concentrations can induce cell proliferation and prolactin release in anterior pituitary cells in culture and whether these effects are mediated through ERs. Here we show that 10 nM Cd was able to stimulate lactotroph proliferation in anterior pituitary cell cultures from female Wistar rats and also in GH3 lactosomatotroph cell line. Proliferation of somatotrophs and gonadotrophs were not affected by Cd exposure. Cd promoted cell cycle progression by increasing cyclins D1, D3 and c-fos expression. Cd enhanced prolactin synthesis and secretion. Cd E2-like effects were blocked by the pure ERs antagonist ICI 182,780 supporting that Cd acts through ERs. Further, both Cd and E2 augmented full-length ERαexpression and its 46 kDa-splicing variant. In addition, when co-incubated Cd was shown to interact with E2 by inducing ERα mRNA expression which indicates an additive effect between them. This study shows for the first time that Cd at nanomolar concentration displays xenoestrogenic activities by inducing cell growth and stimulating prolactin secretion from anterior pituitary cells in an ERs-dependent manner. Cd acting as a potent xenoestrogen can play a key role in the aetiology of different pathologies of the anterior pituitary and in estrogen-responsive tissues which represent considerable risk to human health.


Subject(s)
Cadmium/pharmacology , Lactotrophs/drug effects , Lactotrophs/metabolism , Prolactin/metabolism , Animals , Cadmium/metabolism , Cadmium Chloride/pharmacology , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D3/genetics , Cyclin D3/metabolism , Drug Synergism , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation/drug effects , Prolactin/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
9.
PLoS One ; 7(7): e41299, 2012.
Article in English | MEDLINE | ID: mdl-22844453

ABSTRACT

Estrogens are key to anterior pituitary function, stimulating hormone release and controlling cell fate to achieve pituitary dynamic adaptation to changing physiological conditions. In addition to their classical mechanism of action through intracellular estrogen receptors (ERs), estrogens exert rapid actions via cell membrane-localized ERs (mERs). We previously showed that E2 exerts a rapid pro-apoptotic action in anterior pituitary cells, especially in lactotropes and somatotropes, through activation of mERs. In the present study, we examined the involvement of mERα in the rapid pro-apoptotic action of estradiol by TUNEL in primary cultures of anterior pituitary cells from ovariectomized rats using a cell-impermeable E2 conjugate (E2-BSA) and an ERα selective antagonist (MPP dihydrochloride). We studied mERα expression during the estrous cycle and its regulation by gonadal steroids in vivo by flow cytometry. We identified ERα variants in the plasma membrane of anterior pituitary cells during the estrous cycle and studied E2 regulation of these mERα variants in vitro by surface biotinylation and Western Blot. E2-BSA-induced apoptosis was abrogated by MPP in total anterior pituitary cells and lactotropes. In cycling rats, we detected a higher number of lactotropes and a lower number of somatotropes expressing mERα at proestrus than at diestrus. Acute E2 treatment increased the percentage of mERα-expressing lactotropes whereas it decreased the percentage of mERα-expressing somatotropes. We detected three mERα isoforms of 66, 39 and 22 kDa. Expression of mERα66 and mERα39 was higher at proestrus than at diestrus, and short-term E2 incubation increased expression of these two mERα variants. Our results indicate that the rapid apoptotic action exerted by E2 in lactotropes depends on mERα, probably full-length ERα and/or a 39 kDa ERα variant. Expression and activation of mERα variants in lactotropes could be one of the mechanisms through which E2 participates in anterior pituitary cell renewal during the estrous cycle.


Subject(s)
Cell Membrane/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Lactotrophs/drug effects , Lactotrophs/metabolism , Animals , Apoptosis/drug effects , Cell Membrane/drug effects , Estrous Cycle/drug effects , Female , Lactotrophs/cytology , Rats , Rats, Wistar , Somatotrophs/cytology , Somatotrophs/drug effects , Somatotrophs/metabolism , Time Factors
11.
Endocrinology ; 152(7): 2722-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21521749

ABSTRACT

Dopamine, acting through the dopamine type 2 receptor (Drd2), is the main inhibitor of pituitary prolactin (PRL) secretion and lactotroph proliferation. TGF-ß1 is involved, at least in part, in mediating these actions. It was described that TGF-ß1 synthesis in rat pituitary lactotrophs is up-regulated by dopamine and down-regulated by estradiol. TGF-ß1 is secreted as a large latent complex. The local regulation of cytokine activation in the pituitary has not yet been explored. In this work, we studied pituitary active and total TGF-ß1 content, as well as TGF-ß1 mRNA, and the in vivo role of dopamine and estradiol on pituitary TGF-ß1 levels. Adult female mice (wild type), and female mice with a null mutation in the Drd2 (Drd2(-/-)), were used. The loss of dopaminergic tone induced a decrease in TGF-ß1 mRNA expression, in active and total cytokine content, and in TGF-ß type II receptor expression. Dopamine regulation of pituitary TGF-ß1 activation process was inferred by the inhibition of active cytokine by in vivo sulpiride treatment. Interestingly, in the absence of dopaminergic tone, estradiol induced a strong increase in active TGF-ß1. PRL secretion correlated with active, but not total cytokine. TGF-ß1 inhibitory action on lactotroph proliferation and PRL secretion was decreased in Drd2(-/-) pituitary cells, in correlation with decreased TGF-ß type II receptor. The study of the TGF-ß1 activation process and its regulation is essential to understand the cytokine activity. As an intermediary of dopamine inhibition of lactotroph function, TGF-ß1 and local activators may be important targets in the treatment of dopamine agonist-resistant prolactinomas.


Subject(s)
Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Estradiol/analogs & derivatives , Gene Expression Regulation/drug effects , Pituitary Gland, Anterior/drug effects , Receptors, Dopamine D2/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Proliferation , Cells, Cultured , Dopamine Agonists/therapeutic use , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Hyperprolactinemia/drug therapy , Lactotrophs/drug effects , Lactotrophs/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Prolactin/blood , Prolactin/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics
12.
PLoS One ; 6(3): e18097, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21464994

ABSTRACT

Dopamine, through D2 receptor (D2R), is the major regulator of lactotrope function in the anterior pituitary gland. Both D2R isoforms, long (D2L) and short (D2S), are expressed in lactotropes. Although both isoforms can transduce dopamine signal, they differ in the mechanism that leads to cell response. The administration of D2R agonists, such as cabergoline, is the main pharmacological treatment for prolactinomas, but resistance to these drugs exists, which has been associated with alterations in D2R expression. We previously reported that dopamine and cabergoline induce apoptosis of lactotropes in primary culture in an estrogen-dependent manner. In this study we used an in vivo model to confirm the permissive action of estradiol in the apoptosis of anterior pituitary cells induced by D2R agonists. Administration of cabergoline to female rats induced apoptosis, measured by Annexin-V staining, in anterior pituitary gland from estradiol-treated rats but not from ovariectomized rats. To evaluate the participation of D2R isoforms in the apoptosis induced by dopamine we used lactotrope-derived PR1 cells stably transfected with expression vectors encoding D2L or D2S receptors. In the presence of estradiol, dopamine induced apoptosis, determined by ELISA and TUNEL assay, only in PR1-D2S cells. To study the role of p38 MAPK in apoptosis induced by D2R activation, anterior pituitary cells from primary culture or PR1-D2S were incubated with an inhibitor of the p38 MAPK pathway (SB203850). SB203580 blocked the apoptotic effect of D2R activation in lactotropes from primary cultures and PR1-D2S cells. Dopamine also induced p38 MAPK phosphorylation, determined by western blot, in PR1-D2S cells and estradiol enhanced this effect. These data suggest that, in the presence of estradiol, D2R agonists induce apoptosis of lactotropes by their interaction with D2S receptors and that p38 MAPK is involved in this process.


Subject(s)
Apoptosis/drug effects , Dopamine/pharmacology , Lactotrophs/cytology , Lactotrophs/drug effects , Receptors, Dopamine D2/metabolism , Animals , Cabergoline , Ergolines/pharmacology , Estradiol/pharmacology , Female , Lactotrophs/enzymology , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Neuroendocrinology ; 93(2): 106-13, 2011.
Article in English | MEDLINE | ID: mdl-21252492

ABSTRACT

BACKGROUND: Estrogens are recognized modulators of pituitary cell renewal, sensitizing cells to mitogenic and apoptotic signals. Tumor necrosis factor-α (TNF-α) is a proinflammatory cytokine that plays an important role in tissue homeostasis modulating cell proliferation, differentiation and death. We previously demonstrated that TNF-α-induced apoptosis of anterior pituitary cells from female rats is estrogen-dependent and predominant in cells from rats at proestrus when estradiol levels are the highest. AIMS: Considering that one of the mechanisms involved in the apoptotic action of estrogens can result from increased expression of cytokines and/or their receptors, the aim of the present study was to evaluate the effect of estrogens on the expression of TNF-α and its receptor, TNF receptor 1 (TNFR1), in anterior pituitary cells. METHODS/RESULTS: TNFR1 expression, determined by Western blot, was higher in anterior pituitary glands from rats at proestrus than at diestrus. Incubation of anterior pituitary cells from ovariectomized rats with 17ß-estradiol enhanced TNFR1 protein expression. As determined by double immunocytochemistry, the expression of TNF-α and TNFR1 was detected in prolactin-, GH-, LH- and ACTH-bearing cells. 17ß-estradiol increased the percentage of TNF-α and TNFR1-immunoreactive lactotropes but did not modify the number of GH-bearing cells expressing TNF-α or TNFR1. CONCLUSION: Our results demonstrate that estradiol increases the expression of TNF-α and TNFR1 in anterior pituitary cells, especially in lactotropes. The sensitizing action of estrogens to proapoptotic stimuli at proestrus in the anterior pituitary gland may involve changes in the expression of the TNF-α/TNFR1 system.


Subject(s)
Estradiol/physiology , Gene Expression Regulation/physiology , Lactotrophs/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cells, Cultured , Estradiol/pharmacology , Estrous Cycle/physiology , Female , Lactotrophs/drug effects , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Rats , Rats, Wistar
14.
Exp Physiol ; 96(2): 226-39, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21075822

ABSTRACT

Lactotroph cells display morphological and functional heterogeneity, a feature which is closely related to the oestrogenic environment. In this study, we focused on sex-related differences linked to the proliferative and secretory responses of lactotrophs exposed to EGF in vitro. Furthermore, we addressed the involvement of the PKCε/ERK1/2 signalling pathway and the contribution of Pit-1 in the EGF actions in primary pituitary cultures from male and female rats. EGF promoted a differential proliferative activity on PRL cells, which was closely associated to the sex, as revealed by the uptake of bromodeoxyuridine (BrdU). In females, the mitogenic activity was up to nine times greater, whereas in males, the number of BrdU-labelled PRL cells was only doubled compared to control. However, in both models, EGF had a similar effectiveness in promoting PRL secretion. EGF also induced a significant increase in the PKCε, P -ERK 1/2, and Pit-1 protein levels, which were higher in females than in males. Pre-incubation with BIM blocked EGF-induced ERK 1/2 activation and Pit-1 expression. These results suggest a sexually dimorphic response of lactotroph cells to the proliferative effects of EGF, with the PKCε/ERK1/2 Pit-1 pathway being involved in this action.


Subject(s)
Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , Lactotrophs/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-epsilon/metabolism , Transcription Factor Pit-1/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Lactotrophs/enzymology , Lactotrophs/metabolism , Male , Phosphorylation , Prolactin/metabolism , Rats, Wistar , Sex Characteristics , Sex Factors , Signal Transduction/drug effects
15.
Endocrine ; 39(1): 21-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21069582

ABSTRACT

We have previously reported that Fas activation induces apoptosis of anterior pituitary cells from rats at proestrus but not at diestrus and in an estrogen-dependent manner. In this study, we evaluated the effect of Fas activation on apoptosis of lactotropes and somatotropes during the estrous cycle and explored the action of gonadal steroids on Fas-induced apoptosis. Also, we studied whether changes in Fas expression are involved in the apoptotic response of anterior pituitary cells. Fas activation increased the percentage of TUNEL-positive lactotropes and somatotropes at proestrus but not at diestrus. FasL triggered apoptosis of somatotropes only when cells from ovariectomized rats were cultured in the presence of 17 ß-estradiol (E2). Progesterone (P4) blocked the apoptotic action of the Fas/FasL system in lactotropes and somatotropes incubated with E2. Both E2 and P4 increased the percentage of cells expressing Fas at the cell membrane. Our results show that Fas activation induces apoptosis of lactotropes and somatotropes at proestrus but not at diestrus. Gonadal steroids may be involved in the apoptotic response of lactotropes and somatotropes, suggesting that Fas activation is implicated in the renewal of these pituitary subpopulations during the estrous cycle. The effect of gonadal steroids on Fas expression may be only partially involved in regulation of the Fas/FasL apoptotic pathway in the anterior pituitary gland.


Subject(s)
Apoptosis/drug effects , Gonadal Steroid Hormones/pharmacology , Lactotrophs/cytology , Somatotrophs/cytology , fas Receptor/physiology , Animals , Apoptosis/physiology , Cells, Cultured , Estradiol/pharmacology , Estrous Cycle , Fas Ligand Protein/physiology , Female , Gene Expression/drug effects , In Situ Nick-End Labeling , Lactotrophs/drug effects , Ovariectomy , Pituitary Gland, Anterior/cytology , Progesterone/pharmacology , Rats , Rats, Wistar , Somatotrophs/drug effects , fas Receptor/genetics
17.
Mol Endocrinol ; 23(7): 1102-14, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342445

ABSTRACT

The regulatory role of estrogen, bone morphogenetic protein-4 (BMP-4), and TGF-beta has a strong impact on hormone secretion, gene transcription, and cellular growth of prolactin (PRL)-producing cells. In contrast to TGF-beta, BMP-4 induces the secretion of PRL in GH3 cells. Therefore, we studied the mechanism of their transcriptional regulation. Both BMP-4 and TGF-beta inhibited the transcriptional activity of the estrogen receptor (ER). Estrogens had no effect on TGF-beta-specific Smad protein transcriptional activity but presented a stimulatory action on the transcriptional activity of the BMP-4-specific Smads. BMP-4/estrogen cross talk was observed both on PRL hormone secretion and on the PRL promoter. This cross talk was abolished by the expression of a dominant-negative form for Smad-1 and treatment with ICI 182780 but not by point mutagenesis of the estrogen response element site within the promoter, suggesting that Smad/ER interaction might be dependent on the ER and a Smad binding element. By serial deletions of the PRL promoter, we observed that indeed a region responsive to BMP-4 is located between -2000 and -1500 bp upstream of the transcriptional start site. Chromatin immunoprecipitation confirmed Smad-4 binding to this region, and by specific mutation and gel shift assay, a Smad binding element responsible site was characterized. These results demonstrate that the different transcriptional factors involved in the Smad/ER complexes regulate their transcriptional activity in differential ways and may account for the different regulatory roles of BMP-4, TGF-beta, and estrogens in PRL-producing cells.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Estrogens/pharmacology , Lactotrophs/metabolism , Prolactin/genetics , Promoter Regions, Genetic , Transforming Growth Factor beta/metabolism , Animals , Binding Sites , Bone Morphogenetic Protein 4/physiology , Cells, Cultured , Estrogens/metabolism , Lactotrophs/drug effects , Prolactin/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Rats , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Smad Proteins/metabolism , Smad Proteins/physiology , Transcriptional Activation/physiology , Transforming Growth Factor beta/physiology
18.
J Mol Histol ; 40(5-6): 417-25, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20177957

ABSTRACT

The variations of the intracellular localization of the individual protein kinase C (PKC) isoforms are related with their different biological functions. In this study, we have investigated the precise intracellular translocation of endogenous PKCalpha and PKCepsilon in PMA-stimulated normal and tumoral lactotroph cells by using confocal and immunogold electron microscopy, which was correlated with the rate of cell proliferation of both pituitary cell phenotypes. The present results showed that the short phorbol ester incubation stimulated the proliferation of normal and tumoral lactotroph cells, as determined by the measurement of the BrdU-labelling index. The translocation of PKCalpha to plasma and nuclear membranes induced by PMA was more marked than that observed for PKCepsilon in normal and tumoral lactotroph cells. Our results showed that PKCs translocation to the plasma and nuclear membranes varied from isozyme to isozyme emphasizing that PKCalpha could be related with the mitogenic stimulus exerted by phorbol ester. These data support the notion that specific PKC isozymes may exert spatially defined effects by virtue of their directed translocation to distinct intracellular sites.


Subject(s)
Lactotrophs/enzymology , Lactotrophs/pathology , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Lactotrophs/drug effects , Lactotrophs/ultrastructure , Mitogens/pharmacology , Nuclear Envelope/drug effects , Nuclear Envelope/enzymology , Nuclear Envelope/ultrastructure , Pituitary Neoplasms/enzymology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/ultrastructure , Protein Kinase C-alpha/ultrastructure , Protein Kinase C-epsilon/ultrastructure , Protein Transport/drug effects , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology
19.
Neuroendocrinology ; 89(2): 200-9, 2009.
Article in English | MEDLINE | ID: mdl-18832805

ABSTRACT

BACKGROUND/AIMS: The antiprogesterone mifepristone facilitates prolactin release, an effect enhanced by administration of the opioid antagonist naloxone. The present study explores ultrastructural changes in lactotropes after mifepristone and naloxone administration, correlating them with the expression of pituitary prolactin. METHODS/RESULTS: Rats were sacrificed at 18:00 h on day 19 of pregnancy. Prolactin immunoelectron microscopy of lactotropes from control rats showed characteristics of quiescent cells with numerous small and spherical secretory granules. Naloxone administration did not modify lactotrope morphology or prolactin expression in terms of mRNA or protein abundances. Mifepristone treatment induced lactotrope activation with development of the rough endoplasmic reticulum and Golgi complex with prolactin immunoreactive small newly formed and large mature secretory granules. Mifepristone increased prolactin mRNA and protein expression. Naloxone administration to mifepristone-treated rats potentiated lactotrope activation compared with mifepristone alone showing exocytotic images of prolactin granules and some cells with evident signs of involution. CONCLUSIONS: (1) Blockade of progesterone action by mifepristone activated the lactotrope, increased significantly prolactin mRNA and protein expression and prepared the pituitary for naloxone action. (2) The high serum prolactin levels induced by mifepristone and naloxone may regulate negatively lactotrope activity as suggested by the presence of regressing cells neighboring the actively secreting cells.


Subject(s)
Lactotrophs/drug effects , Lactotrophs/ultrastructure , Mifepristone/agonists , Naloxone/agonists , Pituitary Gland/drug effects , Pituitary Gland/ultrastructure , Prolactin/drug effects , Prolactin/metabolism , Animals , Female , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL