Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Hum Reprod ; 20(11): 1054-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25180269

ABSTRACT

Sperm capacitation involves an increase in intracellular Ca(2+) concentration as well as in protein kinase A (PKA)-dependent protein tyrosine (Tyr) phosphorylation. Interestingly, in humans, a decrease in extracellular Ca(2+) concentration ([Ca(2+)]e) during capacitation induces an increase in Tyr phosphorylation indicating the complexity of Ca(2+) signaling during this process. In view of this, in the present study we further investigated the Ca(2+)-mediated signaling pathways implicated in Tyr phosphorylation during human sperm capacitation. Results revealed that sperm incubation in a medium without added Ca(2+) (⊖ Ca(2+)) increased Tyr phosphorylation but did not modify PKA-mediated phosphorylation. Moreover, inhibition of either PKA or Src family kinase signaling cascades in ⊖ Ca(2+) down-regulated both PKA substrate and Tyr phosphorylations, indicating that the [Ca(2+)]e effects on Tyr phosphorylation depend on PKA targets. Inhibition of calmodulin or Ser/Thr protein phosphatase 2B also increased Tyr phosphorylation without affecting PKA-mediated phosphorylation, supporting the potential role of these Ca(2+) downstream effectors in the increase in Tyr phosphorylation observed in ⊖ Ca(2+). Experiments aimed to identify the kinase responsible for these observations revealed the presence of proline-rich tyrosine kinase 2 (PYK2), a focal adhesion kinase (FAK) family member, in human sperm, and the use of PF431396, an FAK inhibitor, supported the involvement of PYK2 in Tyr phosphorylation downstream of PKA activation. Results also showed that PYK2 was activated in ⊖ Ca(2+) as well as during capacitation and that PF431396 affected capacitated sperm motility, acrosome reaction and ability to penetrate both mouse cumulus matrix and zona-free hamster eggs. Together, our observations support PYK2 as an intermediary component of Ca(2+) signaling between PKA-mediated and Tyr phosphorylations that is required for achieving functional human sperm capacitation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Focal Adhesion Kinase 2/physiology , Sperm Capacitation/physiology , Tyrosine/metabolism , Calcium Signaling , Enzyme Activation , Focal Adhesion Kinase 2/metabolism , Humans , Phosphorylation
2.
Mol Hum Reprod ; 19(9): 570-80, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23630234

ABSTRACT

In all mammalian species studied so far, sperm capacitation correlates with an increase in protein tyrosine (Tyr) phosphorylation mediated by a bicarbonate-dependent cAMP/protein kinase A (PKA) pathway. Recent studies in mice revealed, however, that a Src family kinase (SFK)-induced inactivation of serine/threonine (Ser/Thr) phosphatases is also involved in the signaling pathways leading to Tyr phosphorylation. In view of these observations and with the aim of getting a better understanding of the signaling pathways involved in human sperm capacitation, in the present work we investigated the involvement of both the cAMP/PKA and SFK/phosphatase pathways in relation to the capacitation state of the cells. For this purpose, different signaling events and sperm functional parameters were analyzed as a function of capacitation time. Results revealed a very early bicarbonate-dependent activation of PKA indicated by the rapid (1 min) increase in both phospho-PKA substrates and cAMP levels (P < 0.05). However, a complete pattern of Tyr phosphorylation was detected only after 6-h incubation at which time sperm exhibited the ability to undergo the acrosome reaction (AR) and to penetrate zona-free hamster oocytes. Sperm capacitated in the presence of the SFK inhibitor SKI606 showed a decrease in both PKA substrate and Tyr phosphorylation levels, which was overcome by exposure of sperm to the Ser/Thr phosphatase inhibitor okadaic acid (OA). However, OA was unable to induce phosphorylation when sperm were incubated under PKA-inhibitory conditions (i.e. in the absence of bicarbonate or in the presence of PKA inhibitor). Moreover, the increase in PKA activity by exposure to a cAMP analog and a phosphodiesterase inhibitor did not overcome the inhibition produced by SKI606. Whereas the presence of SKI606 during capacitation produced a negative effect (P < 0.05) on sperm motility, progesterone-induced AR and fertilizing ability, none of these inhibitions were observed when sperm were exposed to SKI606 and OA. Interestingly, different concentrations of inhibitors were required to modulate human and mouse capacitation revealing the species specificity of the molecular mechanisms underlying this process. In conclusion, our results describe for the first time the involvement of both PKA activation and Ser/Thr phosphatase down-regulation in functional human sperm capacitation and provide convincing evidence that early PKA-dependent phosphorylation is the convergent regulatory point between these two signaling pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Phosphoprotein Phosphatases/genetics , Sperm Capacitation/genetics , Spermatozoa/enzymology , src-Family Kinases/genetics , Acrosome Reaction/drug effects , Aniline Compounds/pharmacology , Animals , Cricetinae , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Expression Regulation , Humans , Male , Mice , Nitriles/pharmacology , Okadaic Acid/pharmacology , Oocytes/physiology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Progesterone/pharmacology , Quinolines/pharmacology , Signal Transduction , Sperm Capacitation/drug effects , Sperm Count , Sperm Motility/drug effects , Spermatozoa/cytology , Spermatozoa/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
3.
Mol Hum Reprod ; 12(12): 781-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17050774

ABSTRACT

To become fertilization competent, mammalian sperm undergo changes in the female reproductive tract termed capacitation. Capacitation correlates with an increase in tyrosine phosphorylation; however, less is known about the role of serine/threonine phosphorylation in this process. Proline-directed phosphorylation is one of the major regulatory phosphorylation events in many cellular processes such as cell proliferation and differentiation. Using mitotic phosphoprotein monoclonal-2 (MPM-2) antibody in this study, we observed that several mouse sperm proteins in the range of 70-250 kDa underwent increased serine/threonine-proline phosphorylation during capacitation. In contrast to the time course of tyrosine phosphorylation, proline-directed phosphorylation could be observed at shorter time points of sperm incubation, and it was found to be independent of NaHCO(3) and adenosine 3'5'-cyclic monophosphate (cAMP). Similar to the regulation of the increase in tyrosine phosphorylation, cholesterol acceptors such as bovine serum albumin (BSA) or 2-hydroxypropyl-beta-cyclodextrin (2-OH-propyl-beta-CD) were essential for the regulation of proline-directed phosphorylation in mouse sperm. Furthermore, it was also found to be BSA dependent in human sperm. Among proline-directed kinases, extracellular signal-regulated kinase 1/2 (ERK1/2) is present in mammalian sperm; nevertheless, U0126 and PD098059, two inhibitors of the ERK pathway, did not block this phosphorylation in mouse sperm. In conclusion, capacitation is associated with an increase in proline-directed phosphorylation linked to cholesterol efflux in the sperm.


Subject(s)
Cholesterol/metabolism , Proline/physiology , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/physiology , Sperm Capacitation/physiology , Spermatozoa/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , 2-Hydroxypropyl-beta-cyclodextrin , Amino Acid Motifs , Animals , Antibodies, Monoclonal/immunology , Blotting, Western , Bucladesine/pharmacology , Butadienes/pharmacology , Cattle , Cyclic AMP/agonists , Flavonoids/pharmacology , Humans , Male , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitosis , Nitriles/pharmacology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Serum Albumin, Bovine/pharmacology , Sodium Bicarbonate/pharmacology , Sperm Capacitation/drug effects , Spermatozoa/drug effects , beta-Cyclodextrins/pharmacology
4.
Neuroendocrinology ; 72(5): 272-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11124584

ABSTRACT

The zona incerta (ZI), an area in the dorsal hypothalamus, contains neuronal systems that appear to control gonadotropin release. Previous findings show that there is an inverse relationship between serotonin (5-HT) activity in the ZI and plasma luteinizing hormone (LH) levels, indicating that the 5-HT system in this area has an inhibitory effect on LH release. Employing anaesthetised, ovariectomised rats primed with 5 microg oestradiol benzoate followed at 48 h by 0.5 mg progesterone, we have shown that 2 microg/side 5-HT in the ZI inhibits the LH surge that normally occurs 4 h after the progesterone treatment. This effect was mimicked by 2 microg/side 8-OH-DPAT, a 5-HT1A agonist, but not by DOI, a 5-HT2 agonist, BMY7378, a presynaptic 5-HT1A agonist or MCPP, a 2B & 2C agonist. The inhibitory effect of 5-HT and 8-OH-DPAT was prevented by pretreatment, 1 h before, with either 2 mg/kg i.p. WAY100135, a 5-HT1A antagonist or 0.25 mg/kg i.p. ritanserin, a 5-HT2 antagonist. These results indicate that 5-HT in the ZI exerts its inhibitory effect on LH release via 5-HT1A receptors but that another 5-HT subtype may also be involved.


Subject(s)
Luteinizing Hormone/metabolism , Ovariectomy , Receptors, Serotonin/physiology , Serotonin/pharmacology , Subthalamus/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Anesthesia , Animals , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Piperazines/pharmacology , Progesterone/pharmacology , Rats , Rats, Wistar , Receptors, Serotonin, 5-HT1 , Ritanserin/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Subthalamus/drug effects
5.
Genomics ; 69(1): 54-62, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11013075

ABSTRACT

The OVCA1 gene is a candidate for the breast and ovarian tumor suppressor gene at chromosome 17p13.3. To help determine the function(s) of OVCA1, we used a yeast two-hybrid screening approach to identify OVCA1-associating proteins. One such protein, which we initially referred to as BOV-1 (binder of OVCA1-1) is 173 or 174 amino acids in length and appears to be a new member of a highly conserved RNA-binding motif (RBM) protein family that is highly conserved evolutionarily. Northern blot analysis revealed that BOV-1 is ubiquitously expressed and that three distinct messenger RNA species are expressed, 1-, 3.2-, and 5.8-kb transcripts. The 1-kb transcript is the most abundant and is expressed at high levels in the testis, heart, placenta, spleen, thymus, and lymphocytes. Using fluorescence in situ hybridization and the 5.8-kb complementary DNA probe, we determined that BOV-1 maps to both chromosome 5q13-q14 and chromosome 14q22-q23. Further sequence analysis determined that the gene coding the 1- and the 3.2-kb transcripts (HGMW-approved gene symbol RBM8A) maps to 14q22-q23, whereas a second highly related gene coding for the 5.8-kb transcript resides at chromosome 5q13-q14 (HGMW-approved gene symbol RBM8B). The predicted proteins encoded by RBM8A and RBM8B are identical except that RBM8B is 16 amino acids shorter at its N-terminus. Molecular modeling of the RNA-binding domain of RBM8A and RBM8B, based on homology to the sex-lethal protein of Drosophila, identifies conserved residues in the RBM8 protein family that are likely to contact RNA in a protein-RNA complex. The conservation of sequence and structure through such an evolutionarily divergent group of organisms suggests an important function for the RBM8 family of proteins.


Subject(s)
RNA-Binding Proteins/genetics , Tumor Suppressor Proteins , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Chromosome Mapping , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 5/genetics , Cloning, Molecular , Conserved Sequence , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Female , Gene Expression , Genes, Tumor Suppressor , Humans , In Situ Hybridization, Fluorescence , Male , Microscopy, Fluorescence , Minor Histocompatibility Antigens , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
6.
Int J Oncol ; 16(2): 231-40, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10639564

ABSTRACT

The mechanism of cell immortalization of human breast epithelial cells leading to neoplastic transformation is not clear. The isolation and characterization of a spontaneously immortalized human breast epithelial cell line, MCF-10F, have provided a valuable tool to identify genes involved in this process. Using the technique of differential display, we have identified seven cDNA bands differentially displayed in the MCF-10F cells when compared with the mortal S130 cells from which MCF-10F was originated. One of these bands was isolated and cloned. Sequence analysis revealed 99% homology to the EF-hand calcium-binding protein S100P (Placental). The clone was overexpressed in the immortal cell line MCF-10F when compared to the mortal counterpart S130 or other primary cultures of human breast epithelial cells. In addition, it was highly expressed in chemically transformed breast epithelial cell lines (BP1E and D3. 1), breast cancer cell line T47D, as well as in three invasive ductal carcinomas when compared to their normal adjacent tissue. The S100P protein was localized by immunohistochemistry, using a monoclonal antibody against the same amino acid sequence of the gene cloned, in ductal hyperplasias, in situ and invasive ductal carcinoma, but not in the normal tissues. We concluded that S100P overexpression is an early event that might play an important role in the immortalization of human breast epithelial cells in vitro and tumor progression in vivo.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Calcium-Binding Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Neoplasm Proteins/metabolism , Base Sequence , Breast/pathology , Breast Neoplasms/pathology , Carcinoma in Situ/metabolism , Carcinoma, Ductal, Breast/metabolism , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Female , Humans , Molecular Sequence Data , RNA/metabolism , Tumor Cells, Cultured
7.
Int J Oncol ; 13(1): 65-71, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9625804

ABSTRACT

Cellular proliferation, essential for normal development, may result in neoplastic growth when the cell cycle clock is disrupted. In order to determine whether the protein expression of cell cycle regulators differs among normal, immortalized non-tumorigenic and malignant human breast epithelial cells (HBECs), we analyzed the protein expression of cyclins D1, D3, A and E, cyclin-dependent kinase (cdk) 4 and c-fos in exponentially growing MCF-10M, MCF-10F, and MCF-7 cells. The tumorigenicity of HBECs in vivo correlated with both cell cycle regulators and early-gene protein expression in vitro. The differential expression of cyclin E- and cyclin A-related proteins and their putative relevance in the tumorigenic properties of HBECs are also discussed.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Cell Cycle Proteins/physiology , Cell Transformation, Neoplastic , Cell Division , Cells, Cultured , Epithelial Cells/cytology , Female , Humans , Phenotype , Tumor Cells, Cultured
8.
Eur J Pharmacol ; 332(3): 283-7, 1997 Aug 13.
Article in English | MEDLINE | ID: mdl-9300262

ABSTRACT

It has been shown that adrenal progesterone plays an important role in regulating the negative feedback of oestrogen on luteinizing hormone (LH) release in ovariectomized and oestrogen-treated rats. The purpose of the present study was to determine whether adrenal progesterone modulation of LH secretion is mediated by adrenergic and opioidergic systems in ovariectomized and oestrogen-treated rats. Oestradiol benzoate (20 micrograms/rat) was given s.c. to ovariectomized rats on day 0. Control animals were injected with the vehicle alone. The specific adrenoceptor antagonists prazosin (10 mg/kg), idazoxan (100 micrograms/kg), metoprolol (10 mg/kg) or ICI 118,551 (200 micrograms/kg) were injected at 12.00 and 20.00 h on day 2 and at 08.00 h on day 3 to oestrogen-primed rats treated or not with RU486. Control animals were injected with saline. RU486 (10 mg/kg) was administered s.c. at 08.00 h on day 3 to oestradiol-treated animals receiving adrenoceptor antagonists or saline. Naloxone (2 mg/kg) was administered i.p. 30 min before blood-sampling to oestrogen-primed rats treated or not with RU486. All groups were blood-sampled at 13.00 and 18.00 h on day 3, and LH concentration was measured by radioimmunoassay. The administration of oestradiol to ovariectomized rats decreased serum LH levels at 13.00 and 18.00 h on day 3. Prazosin or idazoxan partially prevented the effect of oestradiol at 13.00 h, while metoprolol, ICI 118,551 or naloxone totally blocked the inhibitory effect of oestradiol on LH secretion; both adrenoceptor and opioid receptor antagonists also prevented the effect of oestrogen on LH concentration at 18.00 h. RU486 increased serum LH concentration at 18.00 h in oestrogen-primed rats to values higher than in ovariectomized control rats, with no effect at noon. The administration of prazosin to ovariectomized and oestrogen-primed rats treated with RU486 prevented this increase while the other adrenoceptor antagonists or naloxone increased serum LH concentrations at 18.00 h. The present study shows that RU486 switches the feedback of oestradiol on LH secretion from negative to positive in ovariectomized and oestradiol-primed rats, activating a stimulatory alpha 1-adrenergic pathway during the afternoon, and gives strong evidence about the participation of adrenal progesterone modulating neurotransmitter systems involved in the secretion of LH. It also supports the participation of endogenous opioid peptides in the negative feedback of oestradiol, suggesting that the inhibitory tone of endogenous opioid peptide is active regardless the action of adrenal progesterone.


Subject(s)
Adrenal Glands/physiology , Luteinizing Hormone/metabolism , Progesterone/physiology , Receptors, Adrenergic/physiology , Receptors, Opioid/physiology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Antagonists , Adrenergic beta-Antagonists/pharmacology , Animals , Estradiol/pharmacology , Feedback/drug effects , Female , Hormone Antagonists/pharmacology , Idazoxan/pharmacology , Luteinizing Hormone/blood , Metoprolol/pharmacology , Mifepristone/pharmacology , Naloxone/pharmacology , Ovariectomy , Prazosin/pharmacology , Propanolamines/pharmacology , Rats , Rats, Wistar
9.
Cancer Res ; 57(3): 378-81, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012459

ABSTRACT

Tamoxifen (TAM) exerts a long-term suppressive effect on human breast cancer cell proliferation. To determine whether the effects of TAM are mediated by specific gene activation or repression, normal and tumoral human breast tissues obtained before and during TAM treatment were analyzed by differential display technique. Total RNA for differential display analysis was obtained from breast tissues from two women with the diagnosis of estrogen receptor-positive stage II (T2N1M0) infiltrating ductal carcinoma, made by incisional biopsy, followed by modified radical mastectomy performed after a 30-day treatment with TAM (20 mg/day). One 202-bp cDNA band, AP5-1, was present in normal and tumoral biopsy samples, but was absent in breast tissue obtained during TAM treatment, and was confirmed by Northern hybridization, which showed a 2.7-kb band in both patients. The differentially expressed cDNA fragment showed 99% homology to Homo sapiens CD36 gene, a glycoprotein that acts as a receptor for the extracellular matrix proteins thrombospondin-1, collagen types I and IV, and oxidized low-density lipoprotein. These results indicate that the down-regulation of CD36 induced by TAM might represent alternative or additional mechanisms of action of this drug affecting the functions of thrombospondin-1, which is involved in hematogenous tumor spread, invasion and angiogenesis, and oxidized low-density lipoprotein, playing a role in inhibition of arteriosclerosis. The multiple functions affected by the down-regulation of CD36 by TAM warrant the need for additional studies.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Breast/drug effects , CD36 Antigens/genetics , Gene Expression Regulation/drug effects , RNA, Messenger/analysis , Tamoxifen/pharmacology , Base Sequence , Breast/metabolism , Breast Neoplasms/drug therapy , Down-Regulation , Female , Humans , Molecular Sequence Data , Transcriptional Activation
10.
Mol Carcinog ; 20(4): 332-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9433477

ABSTRACT

Subtractive hybridization was used to isolate genes expressed uniquely in the immortalized human breast epithelial cell (HBEC) line MCF-10F and not in the mortal HBEC line S-130, from which MCF-10F cells were derived. We identified a 233-bp cDNA that was expressed in MCF-10F cells and not in their mortal counterpart S-130 cells. Sequence comparison with the GenBank database revealed that the cDNA was identical to the gene encoding human ferritin heavy H chain. Northern blot analysis using the isolated cDNA as a probe showed a differentially expressed 1.1-kb transcript of ferritin H in total RNA from the immortal MCF-10F cells, MCF-10F cells treated with the chemical carcinogens 7,12-dimethylbenz[a]anthracene and benzo[a]pyrene, and the breast cancer cell lines MCF-7, HBL-100, T-47D, and BT-20. No ferritin H transcript was detected in the mortal line S-130 or in other primary HBEC cultures. Increased levels of mRNA transcript signals were also detected in total RNA from breast cancer tissue samples. Tissue with ductal hyperplasia had higher expression levels than normal adjacent mammary tissue. In situ hybridization showed high levels of ferritin H transcript in mammary tissue areas with ductal hyperplasia, carcinoma in situ, and infiltrating ductal carcinoma. This is the first report of the differential expression and upregulation of human ferritin H chain gene in immortal HBECs. It may be an important factor in the process of immortalization, possibly an early stage of malignant transformation of HBECs, providing cells with iron necessary for growth and clonal expansion. Also, ferritin iron, once released, may increase the level of reactive iron, leading to an increase in oxygen free-radical generation, oxidative DNA damage, and mutation.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Ferritins/biosynthesis , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cells, Cultured , Cloning, Molecular , DNA, Complementary , Epithelial Cells/metabolism , Female , Ferritins/genetics , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Gene Library , Humans , Models, Biological , Molecular Sequence Data , Neoplasm Invasiveness , Transcription, Genetic , Tumor Cells, Cultured
11.
Neuropharmacology ; 36(10): 1433-8, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9423931

ABSTRACT

The purpose of the present study was to determine whether glucocorticoid inhibition of prolactin (PRL) release in oestrogen-treated ovariectomized (OVX) rats is mediated by endogenous opioid peptides (EOPs). All the animals were OVX and given oestradiol benzoate (OB, 20 microg/rat, s.c.) 2 weeks later (day 0). On day 3 they received vehicle, mifepristone (MIF, 10 mg/kg, s.c.) or hydrocortisone (HYD, 2 mg/rat, s.c.), in combination with the opioid antagonist naloxone (NAL, 2 mg/kg, i.p.) or vehicle. Serum PRL concentration was then measured by RIA at 13.00 and 18.00 hr, to include assessment of diurnal variation of PRL secretion. At 13.00 hr either MIF or NAL alone increased PRL secretion with no additional effect when NAL was combined with MIF. HYD had no significant inhibitory effect, but NAL with HYD increased PRL secretion. At 18.00 hr serum PRL concentration was higher than at 13.00 hr, and not affected significantly by MIF or NAL alone, although PRL secretion was increased by treatment with both. HYD inhibited PRL secretion and this inhibition was prevented by NAL. In a second experiment to distinguish antiglucocorticoid and antiprogesterone effects of MIF, we administered progesterone (2 mg/rat, s.c.) or a specific progesterone antiserum. In contrast with MIF, the progesterone antibody had no effect on PRL secretion at 13.00 hr, nor on the stimulation by NAL, while progesterone (unlike HYD) increased PRL secretion and NAL attenuated this response; this was opposite to the effect of NAL with HYD. Similarly, at 18.00 hr the interaction of MIF and NAL was not explained by antagonism of progesterone. Together, these results indicate inhibition of PRL by glucocorticoids but not progesterone, mediated in part by EOPs. At 18.00 hr endogenous glucocorticoids do not regulate oestrogen-stimulated PRL release, although HYD is inhibitory through EOPs.


Subject(s)
Estradiol/pharmacology , Opioid Peptides/metabolism , Ovariectomy , Prolactin/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Estradiol/administration & dosage , Female , Hormone Antagonists/pharmacology , Hydrocortisone/pharmacology , Mifepristone/pharmacology , Naloxone/pharmacology , Progesterone/immunology , Progesterone/pharmacology , Prolactin/blood , Prolactin/metabolism , Rats , Rats, Wistar/surgery
12.
J Steroid Biochem Mol Biol ; 48(4): 385-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8142316

ABSTRACT

Accumulated evidence indicates that the adrenal cortex is able to regulate prolactin (PRL) secretion in rats. The aim of this study was to determine the participation of adrenal steroids on the regulation of PRL release in ovariectomized (OVX) and oestrogen-treated rats, by using mifepristone or a specific progesterone antiserum. Blood samples were obtained at 13:00 and 18:00 h 3 days after priming with oestradiol benzoate (OB). A significant increase in serum PRL at 13:00 and 18:00 h was induced by OB treatment. The administration of mifepristone to OVX and oestrogen-primed rats enhanced serum PRL increase at 13:00 h, without modifying the values at 18:00 h; while the administration of progesterone antiserum did not modify PRL levels, indicating that the effect of mifepristone on PRL secretion is due to its antiglucocorticoid action. Adrenalectomy induced a release of PRL at 13:00 h similar to that observed in the OVX and oestrogen-primed rats after mifepristone administration. Treatment with a low dose of progesterone (0.1 mg/rat) to OVX, adrenalectomized and oestrogen-primed rats did not modify the effect of adrenalectomy in serum PRL. Progesterone (2 mg/rat) given at 08:00 h to OVX and oestrogen-primed rats increased serum PRL 5 h later. Mifepristone treatment partially reverted the PRL increase induced by progesterone. These results suggest that after a previous sensitization of the pituitary by oestrogen, circulating glucocorticoids may exert a direct inhibitory effect on PRL release. This inhibition takes place at 13:00 h on day 3. On the other hand, the lack of effect of mifepristone or adrenalectomy on the PRL release at 18:00 h may also indicate that neither progesterone nor glucocorticoids modify PRL release induced by oestrogen at this time.


Subject(s)
Adrenal Cortex/physiology , Estrogens/pharmacology , Glucocorticoids/physiology , Mifepristone/pharmacology , Ovariectomy , Prolactin/metabolism , Adrenal Cortex/drug effects , Adrenalectomy , Animals , Estradiol/pharmacology , Female , Immune Sera/pharmacology , Kinetics , Progesterone/immunology , Progesterone/pharmacology , Progesterone/physiology , Rats , Rats, Wistar
13.
J Endocrinol ; 139(2): 253-8, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8308461

ABSTRACT

There is evidence that the adrenals play a role in the regulation of the synthesis and release of gonadotrophins in various vertebrates. The aim of this study was to determine the part played by adrenal steroids, with special reference to progesterone, on the concentration of LH in ovariectomized (OVX) and oestrogen-primed rats. OVX rats received a single s.c. injection of vehicle or oestradiol benzoate (OB, 20 micrograms/rat). This day was designated as day 0. Three or four days later (day 3-day 4), the rats were treated with mifepristone (10 mg/kg) or with two doses of progesterone antiserum and blood samples were obtained at 13.00 and 18.00 h. OB treatment of OVX rats reduced serum LH at 13.00 h and 18.00 h on day 3 but only at 13.00 h on day 4. The administration of mifepristone at 08.00 h to OVX and oestrogen-treated rats induced a significant increase in serum LH at 18.00 h on days 3 and 4, without modifying the values at 13.00 h. When mifepristone was given at 13.00 h a much larger increase in serum LH was obtained at 18.00 h. In OVX and oestrogen-treated rats, adrenalectomy on day 2 (08.00-09.00 h) induced an increase in serum LH at 18.00 h similar to that observed in the OVX and oestrogen-primed rats after mifepristone treatment. In order to determine the specificity of the effect of mifepristone, a group of OVX and oestrogen-treated rats was injected with progesterone antiserum at 08.00 and 13.00 h on day 3.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Estrogens/physiology , Luteinizing Hormone/metabolism , Progesterone/physiology , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenalectomy , Animals , Estradiol/pharmacology , Feedback , Female , Immune Sera/pharmacology , Luteinizing Hormone/blood , Mifepristone/pharmacology , Ovariectomy , Progesterone/immunology , Progesterone/metabolism , Radioimmunoassay , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...