Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
2.
Front Cell Dev Biol ; 11: 1161813, 2023.
Article in English | MEDLINE | ID: mdl-37082622

ABSTRACT

The epidermal growth factor receptor (EGFR) signaling pathway is one of the main pathways responsible for propagating the luteinizing hormone (LH) signal throughout the cumulus cells and the oocyte. Recently, we have proposed the C-C motif chemokine receptor 2 (CCR2) and its main ligand (monocyte chemoattractant protein-1, MCP1) as novel mediators of the ovulatory cascade. Our previous results demonstrate that the gonadotropins (GNT), amphiregulin (AREG), and prostaglandin E2 (PGE2) stimulation of periovulatory gene mRNA levels occurs, at least in part, through the CCR2/MCP1 pathway, proposing the CCR2 receptor as a novel mediator of the ovulatory cascade in a feline model. For that purpose, feline cumulus-oocyte complexes (COCs) were cultured in the presence or absence of an EGFR inhibitor, recombinant chemokine MCP1, and gonadotropins [as an inducer of cumulus-oocyte expansion (C-OE), and oocyte maturation] to further assess the mRNA expression of periovulatory key genes, C-OE, oocyte nuclear maturation, and steroid hormone production. We observed that MCP1 was able to revert the inhibition of AREG mRNA expression by an EGFR inhibitor within the feline COC. In accordance, the confocal analysis showed that the GNT-stimulated hyaluronic acid (HA) synthesis, blocked by the EGFR inhibitor, was recovered by the addition of recombinant MCP1 in the C-OE culture media. Also, MCP1 was able to revert the inhibition of progesterone (P4) production by EGFR inhibitor in the C-OE culture media. Regarding oocyte nuclear maturation, recombinant MCP1 could also revert the inhibition triggered by the EGFR inhibitor, leading to a recovery in the percentage of metaphase II (MII)-stage oocytes. In conclusion, our results confirm the chemokine receptor CCR2 as a novel intermediate in the ovulatory cascade and demonstrate that the EGFR/AREG and the CCR2/MCP1 signaling pathways play critical roles in regulating feline C-OE and oocyte nuclear maturation, with CCR2/MCP1 signaling pathway being downstream EGFR/AREG pathway within the ovulatory cascade.

3.
Mol Hum Reprod ; 26(5): 289-300, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32159806

ABSTRACT

Expression of immune function genes within follicle cells has been reported in ovaries from many species. Recent work from our laboratory showed a direct effect of the monocyte chemoattractant protein 1/C-C motif chemokine receptor 2 system within the feline cumulus oocyte complex, by increasing the mRNA levels of key genes involved in the ovulatory cascade in vitro. Studies were designed to evaluate if C-C motif chemokine receptor 2 acts as a novel mediator of the ovulatory cascade in vitro. Therefore, feline cumulus oocyte complexes were cultured in the presence or absence of a highly selective C-C motif chemokine receptor 2 antagonist together with known inducers of cumulus-oocyte expansion and/or oocyte maturation to assess mRNA expression of key genes related to periovulatory events in other species as well as oocyte maturation. Also, the effects of recombinant monocyte chemoattractant protein 1 on spontaneous or gonadotrophin-induced oocyte maturation were assessed. This is an in vitro system using isolated cumulus oocyte complexes from feline ovaries. The present study reveals the modulation of several key ovulatory genes by a highly selective C-C motif chemokine receptor 2 antagonist. However, this antagonist was not enough to block the oocyte maturation induced by gonadotropins or amphiregulin. Nonetheless, recombinant monocyte chemoattractant protein 1 had a significant effect on spontaneous oocyte maturation, increasing the percentage of metaphase II stage oocytes in comparison to the control. This is the first study in any species to establish C-C motif chemokine receptor 2 as a mediator of some actions of the mid-cycle gonadotrophin surge.


Subject(s)
Ovulation/genetics , Receptors, CCR2/physiology , Animals , Cats , Cells, Cultured , Cumulus Cells/metabolism , Cumulus Cells/physiology , Female , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/metabolism , Oocytes/physiology , Oogenesis/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/physiology
4.
Horm Metab Res ; 48(4): 275-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26566102

ABSTRACT

Anterior pituitary cell turnover depends on a tight balance between proliferation and apoptosis. We have previously shown that estrogens sensitize anterior pituitary cells to pro-apoptotic stimuli. c-FLIP (cellular-FLICE-inhibitory-protein) isoforms are regulatory proteins of apoptosis triggered by death receptors. c-FLIPshort isoform competes with procaspase-8 inhibiting its activation. However, c-FLIPlong isoform may have a pro- or anti-apoptotic function depending on its expression level. In the present study, we explored whether estrogens modulate c-FLIP expression in anterior pituitary cells from ovariectomized (OVX) rats and in GH3 cells, a somatolactotrope cell line. Acute administration of 17ß-estradiol to OVX rats increased c-FLIPlong expression in the anterior pituitary gland without changing c-FLIPshort expression as assessed by Western blot. Estradiol in vitro also increased c-FLIPlong expression in anterior pituitary cells but not in GH3 cells. As determined by flow cytometry, the percentage of anterior pituitary cells expressing c-FLIP was higher than in GH3 cells. However, c-FLIP fluorescence intensity in GH3 cells was higher than in anterior pituitary cells. FasL increased the percentage of TUNEL-positive GH3 cells incubated either with or without estradiol suggesting that the pro-apoptotic action of Fas activation is estrogen-independent. Our results show that unlike what happens in nontumoral pituitary cells, estrogens do not modulate either c-FLIPlong expression or FasL-induced apoptosis in GH3 cells. The stimulatory effect of estradiol on c-FLIPlong expression could be involved in the sensitizing effect of this steroid to apoptosis in anterior pituitary cells. The absence of this estrogenic action in tumor pituitary cells could be involved in their tumor-like behavior.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Estradiol/metabolism , Pituitary Gland, Anterior/metabolism , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cells, Cultured , Estrogens/metabolism , Female , Pituitary Gland, Anterior/cytology , Rats , Rats, Wistar , Up-Regulation
5.
J Neuroendocrinol ; 27(9): 692-701, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26052658

ABSTRACT

Activation of nuclear factor (NF)-κB promotes cell proliferation and inhibits apoptosis. We have previously shown that oestrogens sensitise normal anterior pituitary cells to the apoptotic effect of tumour necrosis factor (TNF)-α by inhibiting NF-κB nuclear translocation. In the present study, we examined whether oestrogens also modulate the NF-κB signalling pathway and apoptosis in GH3 cells, a rat somatolactotroph tumour cell line. As determined by Western blotting, 17ß-oestradiol (E2 ) (10(-9) m) increased the nuclear concentration of NF-κB/p105, p65 and p50 in GH3 cells. However, E2 did not modify the expression of Bcl-xL, a NF-κB target gene. TNF-α induced apoptosis of GH3 cells incubated in either the presence or absence of E2 . Inhibition of the NF-kB pathway using BAY 11-7082 (BAY) (5 µm) decreased the viability of GH3 cells and increased the percentage of terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL)-positive GH3 cells. BAY also increased TNF-α-induced apoptosis of GH3 cells, an effect that was further increased by an inhibitor of the c-Jun N-terminal protein kinase pathway, SP600125 (10 µm). We also analysed the role of the NF-κB signalling pathway on proliferation and apoptosis of GH3 tumours in vivo. The administration of BAY to nude mice bearing GH3 tumours increased the number of TUNEL-positive cells and decreased the number of proliferating GH3 cells. These findings suggest that GH3 cells lose their oestrogenic inhibitory action on the NF-κB pathway and that the pro-apoptotic effect of TNF-α on these tumour pituitary cells does not require sensitisation by oestrogens as occurs in normal pituitary cells. NF-κB was required for the survival of GH3 cells, suggesting that pharmacological inhibition of the NF-κB pathway could interfere with pituitary tumour progression.


Subject(s)
Apoptosis/physiology , Estrogens/metabolism , Lactotrophs/metabolism , NF-kappa B/metabolism , Pituitary Neoplasms/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Female , Mice , Mice, Nude , Rats , Rats, Wistar
6.
J Neuroendocrinol ; 23(8): 651-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21564349

ABSTRACT

Nuclear factor-kappa B (NF-κB), an important pro-inflammatory factor, is a crucial regulator of cell survival. Both lipopolysaccharide (LPS) and tumour necrosis factor (TNF)-α activate NF-κB signalling. Oestrogens were shown to suppress NF-κB activation. Oestrogens exert a sensitising action to pro-apoptotic stimuli such as LPS and TNF-α in anterior pituitary cells. In the present study, we show by western blotting that 17ß-oestradiol (E(2)) decreases TNF-α-induced NF-κB/p65 and p50 nuclear translocation in primary cultures of anterior pituitary cells from ovariectomised (OVX) rats. Also, the in vivo administration of E(2) decreases LPS-induced NF-κB/p65 and p50 nuclear translocation. To investigate whether the inhibition of NF-κB pathway sensitises anterior pituitary cells to pro-apoptotic stimuli, we used an inhibitor of NF-κB activity, BAY 11-7082 (BAY). BAY, at a concentration that fails to induce apoptosis, has permissive action on TNF-α-induced apoptosis of lactotrophs and somatotrophs from OVX rats, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL). Pharmacological inhibition of NF-κB signalling enhances E(2)-sensitising effect to TNF-α-induced apoptosis in lactotrophs but not in somatotrophs. In vivo administration of BAY allowed LPS-induced apoptosis in anterior pituitary cells from OVX rats (determined by fluorescence activated cell sorting). Furthermore, LPS-induced expression of Bcl-xL in pituitaries of OVX rats is decreased by E(2) administration. Our results show that inhibition of the NF-κB signalling pathway sensitises anterior pituitary cells to the pro-apoptotic action of LPS and TNF-α. Because E(2) inhibits LPS- and TNF-α-activated NF-κB nuclear translocation, the present study suggests that E(2) sensitises anterior pituitary cells to TNF-α- and LPS-induced apoptosis by inhibiting NF-κB activity.


Subject(s)
Apoptosis/drug effects , Lipopolysaccharides/pharmacology , NF-kappa B/antagonists & inhibitors , Pituitary Gland, Anterior/cytology , Tumor Necrosis Factor-alpha/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Cells, Cultured , Estradiol/pharmacology , Female , NF-kappa B/metabolism , Nitriles/pharmacology , Ovariectomy , Rats , Rats, Wistar , Signal Transduction/drug effects , Sulfones/pharmacology
7.
Am J Physiol Endocrinol Metab ; 296(4): E664-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158323

ABSTRACT

It is now accepted that estrogens not only stimulate lactotrope proliferation but also sensitize anterior pituitary cells to proapoptotic stimuli. In addition to their classical mechanism of action through binding to intracellular estrogen receptors (ERs), there is increasing evidence that estrogens exert rapid actions mediated by cell membrane-localized ERs (mERs). In the present study, we examined the involvement of membrane-initiated steroid signaling in the proapoptotic action of estradiol in primary cultures of anterior pituitary cells from ovariectomized rats by using estren, a synthetic estrogen with no effect on classical transcription and a cell-impermeable 17beta-estradiol conjugate (E2-BSA). Both compounds induced cell death of anterior pituitary cells after 60 min of incubation as assessed by flow cytometry and the [3-(4,5-dimethylthiazol-2-yl)]-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Estren, E2, and E2-BSA induced apoptosis of lactotropes and somatotropes as evaluated by the deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay and immunodetection of prolactin (PRL) and growth hormone (GH). The proapoptotic effect of E2-BSA was abrogated by ICI-182,780, an antagonist of ERs. The expression of membrane-associated ERalpha was observed in PRL- and GH-bearing cells. Our results indicate that estradiol is able to exert a rapid apoptotic action in anterior pituitary cells, especially lactotropes and somatotropes, by a mechanism triggered by mERs. This mechanism could be involved in anterior pituitary cell turnover.


Subject(s)
Apoptosis/drug effects , Estrogens/pharmacology , Pituitary Gland, Anterior/drug effects , Animals , Cells, Cultured , Estradiol/pharmacology , Estrenes/pharmacology , Female , Growth Hormone/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/physiology , Prolactin/metabolism , Rats , Rats, Wistar , Receptors, Estrogen/metabolism , Time Factors
8.
Front Horm Res ; 35: 9-21, 2006.
Article in English | MEDLINE | ID: mdl-16809919

ABSTRACT

The anterior pituitary gland undergoes a process of cell renewal during the estrous cycle. Although the occurrence of proliferation and death of anterior pituitary cells at specific stages of the estrous cycle is well known, the underlying mechanisms that regulate these processes are still being uncovered. In spite of the recognized proliferative effects of estrogens on lactotropes, recent evidence shows that estrogens can also trigger antiproliferative and apoptotic responses in anterior pituitary cells. In the present review we analyze the actions of gonadal steroids on proliferation and death of anterior pituitary cells during the estrous cycle and the mediators involved in these actions. Estradiol sensitizes anterior pituitary cells not only to mitogenic stimuli but also to apoptotic signals and upregulates local synthesis of tropic growth factors as well as proapoptotic cytokines. Several growth factors and cytokines have been shown to induce estrogen-dependent lactotrope proliferation and death, whereas progesterone antagonizes estrogen-induced effects. These locally synthesized factors may mediate the effects of gonadal steroids in the process of anterior pituitary cell renewal during the estrous cycle.


Subject(s)
Estrous Cycle/physiology , Pituitary Gland, Anterior/physiology , Animals , Apoptosis/physiology , Cell Death , Cell Proliferation , Humans , Models, Biological
9.
J Endocrinol ; 189(3): 681-90, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731798

ABSTRACT

Our previous work showed that tumor necrosis factor (TNF)-alpha and FasL induce apoptosis of anterior pituitary cells. To further analyze the effect of these proapoptotic factors, we infected primary cultures from rat anterior pituitary, GH3 and AtT20 cells with first-generation adenoviral vectors encoding TNF-alpha, FasL or, as a control, beta-galactosidase (beta-Gal), under the control of the human cytomegalovirus promoter. Successful expression of the encoded transgenes was determined by immunocytochemistry. Although we observed basal expression of TNF-alpha and FasL in control cultures of anterior pituitary cells, fluorescence-activated cell sorting (FACS) cell cycle analysis showed that the overexpression of TNF-alpha or FasL increases the percentage of hypodiploid lactotropes and somatotropes. Nuclear morphology and TUNEL staining revealed that the cells undergo an apoptotic death process. We detected strong immunoreactivity for TNFR1 and Fas in the somatolactotrope cell line GH3. TNF-alpha, but not FasL, was expressed in control cultures of GH3 cells. The infection of GH3 cells with adenovirus encoding TNF-alpha or FasL increased the percentages of hypodiploid and TUNEL-positive cells. TNF-alpha or FasL immunoreactivity was not observed in the corticotrope cell line AtT20. However, adenovirus encoding TNF-alpha or FasL efficiently transduced these cells and increased the percentages of hypodiploid and TUNEL-positive cells. The expression of beta-Gal was detected in all these cultures but did not affect cell viability. In conclusion, these results suggest that death signaling cascades triggered by TNF receptor 1 (TNFR1) and Fas are present in both normal and tumoral pituitary cells. Therefore, overexpression of proapoptotic factors could be a useful tool in the therapy of pituitary adenomas.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/administration & dosage , Membrane Glycoproteins/genetics , Pituitary Gland, Anterior/cytology , Pituitary Neoplasms/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factors/genetics , Animals , Apoptosis , Cell Line, Tumor , Fas Ligand Protein , Female , Flow Cytometry , Gene Expression , Genetic Vectors/genetics , Immunohistochemistry/methods , Membrane Glycoproteins/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Pituitary Neoplasms/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors/metabolism
10.
Endocrinology ; 146(11): 4737-44, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16099864

ABSTRACT

The Fas/FasL system provides the major apoptotic mechanism for many cell types, participating in cell turnover in hormone-dependent tissues. In the present study, we localized both Fas and FasL in anterior pituitary cells, mainly in lactotropes and somatotropes. The percentage of anterior pituitary cells showing immunoreactivity for Fas or FasL was higher in cells from rats killed in proestrus than in diestrus. Also, the proportion of pituitary cells from ovariectomized (OVX) rats expressing Fas or FasL increased in the presence of 17beta-estradiol (10(-9) M). This steroid increased the percentage of lactotropes with immunoreactivity for Fas or FasL and the percentage of somatotropes expressing Fas. Activation of Fas by an agonist anti-Fas antibody (Mab-Fas) decreased the vi-ability-3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT assay)-of anterior pituitary cells from OVX rats cultured in the presence of 17beta-estradiol. Also, membrane-bound FasL decreased cell viability-[3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay (MTS assay)-only when anterior pituitary cells from OVX rats were incubated with 17beta-estradiol. Moreover, FasL increased the percentage of hypodiploid anterior pituitary cells (flow cytometry). Mab-Fas increased the percentage of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL)-positive pituitary cells and lactotropes from OVX rats only when cells were incubated in the presence of 17beta-estradiol. Also, Mab-Fas triggered apoptosis of anterior pituitary cells from rats killed at proestrus but not at diestrus. Our results show that 17beta-estradiol up-regulates the expression of the Fas/FasL system in anterior pituitary cells and increases Fas-induced apoptosis in lactotropes, suggesting that Fas-induced apoptosis could be involved in the pituitary cell renewal during the estrous cycle.


Subject(s)
Apoptosis/physiology , Estrogens/physiology , Membrane Glycoproteins/metabolism , Pituitary Gland, Anterior/physiology , Prolactin/metabolism , Tumor Necrosis Factors/metabolism , fas Receptor/metabolism , Animals , Diestrus , Estradiol/pharmacology , Fas Ligand Protein , Female , Ovariectomy , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Proestrus , Rats , Rats, Wistar , Up-Regulation
11.
Endocrinology ; 146(2): 736-43, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15528300

ABSTRACT

We previously reported that TNF-alpha-induced apoptosis of lactotropes is estrogen dependent and predominant at proestrus. Here we observed that TNF-alpha (50 ng/ml) failed to induce apoptosis of anterior pituitary cells from ovariectomized rats cultured in the presence of progesterone (10(-6) m). However, progesterone blocked the apoptotic effect of TNF-alpha in anterior pituitary cells and lactotropes cultured with 17beta-estradiol (10(-9) m). In addition, 17beta-estradiol induced apoptosis of somatotropes and triggered the proapoptotic action of TNF-alpha in these cells, effects completely blocked by ICI 182 780 (10(-6) m), an estrogen receptor antagonist. Progesterone reverted the permissive effect of 17beta-estradiol on TNF-alpha-induced apoptosis of somatotropes. TNF-alpha induced apoptosis of somatotropes from rats killed at proestrus but not at diestrus. The antiprogestine ZK 98,299 (10(-6) m) completely inhibited the protective action of progesterone on TNF-alpha-induced apoptosis of anterior pituitary cells, lactotropes, and somatotropes. Although progesterone can interact with glucocorticoid receptors, dexamethasone (10(-6) m) had no effect on TNF-alpha-induced apoptosis of anterior pituitary cells, lactotropes, and somatotropes. Our results show that progesterone, by interacting with progesterone receptors, antagonizes the permissive action of estrogens on TNF-alpha-induced apoptosis of lactotropes and somatotropes. These observations suggest that the steroid milieu may modulate the apoptotic response of anterior pituitary cells during the estrous cycle.


Subject(s)
Apoptosis/drug effects , Estradiol/pharmacology , Pituitary Gland, Anterior/cytology , Progesterone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Dexamethasone/pharmacology , Drug Interactions , Female , Glucocorticoids/pharmacology , Ovariectomy , Pituitary Gland, Anterior/drug effects , Rats , Rats, Wistar , Receptors, Progesterone/physiology
12.
Endocrinology ; 145(10): 4677-84, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15208212

ABSTRACT

Glutamate can induce neuronal cell death by activating ionotropic glutamate receptors (iGluRs) as well as metabotropic glutamate receptors (mGluRs). In the present study, we investigated whether glutamate induces apoptosis of cultured anterior pituitary cells from female rats. Glutamate (1 mm) significantly reduced the metabolic activity of viable cells and increased the percentage of terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)-positive cells and caspase-3 activity in anterior pituitary cells. The inhibitory effect of glutamate on the viability of anterior pituitary cells was not observed in the presence of [2S]-alpha-ethylglutamic acid (0.75 mm), a specific group II mGluR antagonist. Also, (2S,1'S,2'S)-2-(carboxycyclopropyl)glycine (LCCG-I; 0.75 mm), a specific group II mGluR agonist, reduced viability and increased the percentage of TUNEL-positive anterior pituitary cells. Group I and III mGluRs and iGluRs agonists failed to modify the metabolic activity of anterior pituitary cells. Glutamate and LCCG-I increased the percentage of TUNEL-positive lactotropes and somatotropes. The subunit mGluR2/3, belonging to group II mGluR, was localized in these cell types. Glutamate increased nitric oxide (NO) synthase (NOS) activity and inducible NOS expression in anterior pituitary cells. N-methyl-l-arginine (NMMA, 0.5 mm), a NOS inhibitor, potentiated the apoptotic effect of glutamate in anterior pituitary cells, indicating that NO may restrain glutamate-induced apoptosis. Incubation of anterior pituitary cells with a cAMP analog (N6, 2'-o-dibutyryladenosine 3', 5'-cyclic monophosphate; 1 mm) attenuated the apoptosis induced by glutamate. Glutamate and LCCG-I decreased prolactin release from anterior pituitary cells. N6, 2'-o-dibutyryladenosine 3', 5'-cyclic monophosphate reversed the inhibitory effect of glutamate on prolactin release, but NMMA failed to modify it. Our data show that glutamate induces apoptosis of lactotropes and somatotropes through group II mGluR activation, probably by decreasing cAMP synthesis.


Subject(s)
Apoptosis/drug effects , Glutamic Acid/pharmacology , Pituitary Gland, Anterior/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Cells, Cultured , Cyclic AMP/physiology , Female , Gene Expression , Nitric Oxide/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Prolactin/metabolism , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Tissue Distribution
13.
Am J Physiol Endocrinol Metab ; 287(4): E767-71, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15172886

ABSTRACT

Tissue homeostasis results from a balance between cell proliferation and cell death by apoptosis. Estradiol affects proliferation as well as apoptosis in hormone-dependent tissues. In the present study, we investigated the apoptotic response of the anterior pituitary gland to lipopolysaccharide (LPS) in cycling female rats, and the influence of estradiol in this response in ovariectomized (OVX) rats. The OVX rats were chronically estrogenized with implanted Silastic capsules containing 1 mg of 17beta-estradiol (E2). Cycling or OVX and E2-treated rats were injected with LPS (250 microg/rat ip). Apoptosis was determined by the terminal deoxynucleotidyl-mediated dUTP nick-end labeling (TUNEL) method in sections of the anterior pituitary gland and spleen. Chronic estrogenization induced apoptosis in the anterior pituitary gland. Acute endotoxemia triggered apoptosis of cells in the anterior pituitary gland of E2-treated rats but not of OVX rats. No differences were observed in the apoptotic response to LPS in spleen between OVX and E2-treated rats. The apoptotic response of the anterior pituitary to LPS was variable along the estrous cycle, being higher at proestrus than at estrus or diestrus I. Approximately 75% of the apoptotic cells were identified as lactotropes by immunofluorescence. In conclusion, our results indicate that estradiol induces apoptosis and enables the proapoptotic action of LPS in the anterior pituitary gland. Also, our study suggests that estrogens may be involved in anterior pituitary cell renewal during the estrous cycle, sensitizing lactotropes to proapoptotic stimuli.


Subject(s)
Apoptosis/drug effects , Estrogens/pharmacology , Pituitary Gland, Anterior/cytology , Animals , Endotoxemia/metabolism , Estradiol/pharmacology , Estrous Cycle/physiology , Female , In Situ Nick-End Labeling , Lipopolysaccharides/pharmacology , Ovariectomy , Pituitary Gland, Anterior/drug effects , Prolactin/metabolism , Rats , Rats, Wistar
14.
Eur J Clin Invest ; 32(11): 818-25, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12423322

ABSTRACT

BACKGROUND: Even if physical activity constitutes a well-known antiatherogenic factor, the precise mechanisms underlying this protective effect are not completely clear. MATERIALS AND METHODS: Lipid and antioxidant profiles were evaluated in 15 well-trained rugby players and 15 sedentary controls. Lipoprotein fractions were separated by sequential ultracentrifugation and alpha-tocopherol content was determined in each fraction by high-performance liquid chromatography. Susceptibility to in vitro oxidation was also measured in intermediate and low density lipoproteins isolated from both groups of subjects as the production of conjugated dienes. RESULTS: Although the sportsmen were not receiving any special diet or vitamin supplementation they showed a slightly improved lipoprotein profile, mainly represented by increased high density lipoprotein-cholesterol levels (P < 0.05), and an enhanced antioxidant status. The latter was evidenced by an increment in total radical antioxidant potential (P < 0.001), higher ascorbic acid (P < 0.005) and alpha-tocopherol (P < 0.05) plasma concentrations, and elevated activities of superoxide dismutase (P < 0.001) and arylesterase (P < 0.01). Moreover, only the fraction of intermediate and low density lipoproteins from rugby players presented higher alpha-tocopherol content in comparison with sedentary controls (484 +/- 67 vs. 377 +/- 123 microg dL(-1), respectively; P < 0.01). Nevertheless, the susceptibility to in vitro oxidation of this lipoprotein fraction was not different between both groups. CONCLUSIONS: Given that intermediate density and low density lipoproteins represent the most atherogenic fraction, this finding, in combination with the improved lipid and antioxidant status, would add to the link between regular physical activity and protection against cardiovascular disease.


Subject(s)
Antioxidants/analysis , Football/physiology , Lipoproteins, LDL/metabolism , Physical Endurance/physiology , Adult , Ascorbic Acid/blood , Carboxylic Ester Hydrolases/metabolism , Case-Control Studies , Catalase/blood , Cholesterol, HDL/blood , Humans , Male , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/analysis , Vitamin E/blood
15.
Endocrinology ; 143(9): 3611-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193577

ABSTRACT

TNF-alpha is involved in the regulation of normal tissue homeostasis affecting cell proliferation, differentiation, and death. We previously reported that TNF-alpha reduces anterior pituitary cell proliferation and PRL release in an estrogen-dependent manner. In the present project we studied the induction of apoptosis by TNF-alpha in anterior pituitary cells from female rats. TNF-alpha (50 ng/ml) decreased the viability of anterior pituitary cells. Incubation with TNF-alpha for 24 h increased the percentage of terminal deoxynucleotidyltransferase-mediated deoxyuridine triphosphate nick end labeling-positive cells. TNF-alpha increased the percentage of somatotropes and lactotropes with apoptotic nuclear morphology without affecting the proportion of apoptotic corticotropes or gonadotropes. TNF-alpha increased the percentage of apoptotic lactotropes in cultured cells from rats killed in proestrus and estrus, but not in diestrus. This effect was significantly higher in cells from rats in proestrus than in estrus. In anterior pituitary cells from ovariectomized rats, TNF-alpha significantly increased the percentage of apoptotic lactotropes only when the cells were incubated in the presence of 17beta-estradiol. These results indicate that TNF-alpha induces apoptosis in somatotropes and lactotropes from female rats. The apoptotic effect of TNF-alpha on lactotropes is dependent on estrogens and could be involved in the regulation of anterior pituitary cell renewal during the estrous cycle.


Subject(s)
Apoptosis , Pituitary Gland, Anterior/cytology , Prolactin/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Electrophoresis, Agar Gel , Estradiol/pharmacology , Estrous Cycle , Estrus , Female , Growth Hormone/metabolism , In Situ Nick-End Labeling , Ovariectomy , Pituitary Gland, Anterior/metabolism , Proestrus , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL