Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
Endocrinology ; 163(3)2022 03 01.
Article in English | MEDLINE | ID: mdl-35085394

ABSTRACT

Prolactin (PRL), a hormone involved in lactation, is mainly produced and secreted by the lactotrophs of the anterior pituitary (AP) gland. We previously reported a method to generate functional adrenocorticotropic hormone-producing cells by differentiating the AP and hypothalamus simultaneously from human induced pluripotent stem cells (iPSCs). However, PRL-producing cells in the induced AP have not been investigated. Here, we confirmed the presence of PRL-producing cells and evaluated their endocrine functions. We differentiated pituitary cells from human iPSCs using serum-free floating culture of embryoid-like aggregates with quick reaggregation (SFEB-q) method and evaluated the appearance and function of PRL-producing cells. Secretion of PRL from the differentiated aggregates was confirmed, which increased with further culture. Fluorescence immunostaining and immunoelectron microscopy revealed PRL-producing cells and PRL-positive secretory granules, respectively. PRL secretion was promoted by various prolactin secretagogues such as thyrotropin-releasing hormone, vasoactive intestinal peptide, and prolactin-releasing peptide, and inhibited by bromocriptine. Moreover, the presence of tyrosine hydroxylase-positive dopaminergic nerves in the hypothalamic tissue area around the center of the aggregates connecting to PRL-producing cells indicated the possibility of recapitulating PRL regulatory mechanisms through the hypothalamus. In conclusion, we generated pituitary lactotrophs from human iPSCs; these displayed similar secretory responsiveness as human pituitary cells in vivo. In the future, this is expected to be used as a model of human PRL-producing cells for various studies, such as drug discovery, prediction of side effects, and elucidation of tumorigenic mechanisms using disease-specific iPSCs. Furthermore, it may help to develop regenerative medicine for the pituitary gland.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Lactotrophs/physiology , Pituitary Gland, Anterior/cytology , Prolactin/biosynthesis , Cell Culture Techniques , Cell Line , Cell Proliferation , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Lactotrophs/drug effects , Prolactin-Releasing Hormone/pharmacology , Thyrotropin-Releasing Hormone/pharmacology , Vasoactive Intestinal Peptide/pharmacology
2.
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
3.
Mol Cell Endocrinol ; 529: 111269, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33819522

ABSTRACT

Prolactin (PRL), mainly synthesized and secreted by the lactotrophs and somatolactotrophs of the anterior pituitary, is a pleiotropic hormone that regulates lactation. In the last decade, nesfatin-1 (NESF) and NESF-like peptide (NLP), encoded in nucleobindin 1 and 2 (NUCB1 and NUCB2), respectively, were characterized as metabolic factors with a potential role in the control of pituitary hormones. We hypothesized that NUCBs and their encoded peptides (NESF and NLP) suppress PRL transcription in the pituitary. The main objective of this research was to determine whether exogenous NESF and NLP, and/or endogenous NUCB1 and NUCB2 regulate the expression of prl and preb mRNAs. Using immortalized rat somatolactotrophs (GH3 cells), dose-response studies were performed to test whether NESF and NLP affect prl and preb. Moreover, the ability of these peptides to modulate the effects of the PRL stimulator thyrotropin releasing hormone (TRH) was studied. Besides, the effects of siRNA-mediated knockdown of endogenous NUCBs on prl and preb mRNAs were determined. NESF and NLP reduced the transcription of prl and preb in GH3 cells. Both NESF and NLP also prevented the stimulatory effects of TRH prl and preb expression. The knockdown of endogenous NUCB1 attenuates both basal prl and TRH-induced expression of prl and preb, while the silencing of NUCBs did not affect the actions of exogenous NESF or NLP. Overall, this work reveals that NUCBs and encoded-peptides are novel regulators of PRL. Future research should test whether the effects observed here in GH3 cells are preserved both in vivo and at the post-transcriptional level.


Subject(s)
DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Lactotrophs/drug effects , Nucleobindins/pharmacology , Peptides/pharmacology , Prolactin/genetics , Somatotrophs/drug effects , Transcription Factors/genetics , Animals , Cell Line, Transformed , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Lactotrophs/cytology , Lactotrophs/metabolism , Nucleobindins/antagonists & inhibitors , Nucleobindins/genetics , Nucleobindins/metabolism , Prolactin/antagonists & inhibitors , Prolactin/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Somatotrophs/cytology , Somatotrophs/metabolism , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
4.
Exp Clin Endocrinol Diabetes ; 129(1): 22-28, 2021 Jan.
Article in English | MEDLINE | ID: mdl-31234219

ABSTRACT

BACKGROUND: Metformin reduced prolactin levels only in women with hyperprolactinemia. OBJECTIVE: The purpose of this case-control study was to compare metformin action on lactoctrope function between women receiving oral contraceptive pills and women not using hormonal contraception. METHODS: The study included two groups of matched women with elevated prolactin levels and new-onset prediabetes or diabetes. The first group consisted of 20 women using oral contraceptive pills for at least 12 months before entering the study, while the second group included 20 patients not using any hormonal contraception. Over the whole study period, all women were treated with metformin (1.7-3 g daily). Circulating levels of glucose, insulin, prolactin, thyrotropin, free thyroid hormones, adrenocorticotropic hormone, gonadotropins and insulin-like growth factor-1 were measured at the beginning and at the end of the study (16 weeks later). RESULTS: Thirty-eight patients completed the study. Metformin reduced plasma glucose levels and improved insulin sensitivity but the latter effect was stronger in women receiving oral contraceptive pills than in women not using any contraception. Although metformin treatment decreased plasma prolactin levels in both study groups, this effect was stronger in women taking oral contraceptive pills. Only in this group of women, metformin increased plasma luteinizing hormone levels. The changes in plasma prolactin correlated with their baseline insulin sensitivity and the effect of metformin on insulin sensitivity. Metformin did not affect plasma levels of thyrotropin, free thyroxine, free triiodothyronine, follicle-stimulating hormone, adrenocorticotropic hormone and insulin-like growth factor-1. CONCLUSIONS: The obtained results suggest that the effect of metformin on overactive lactotropes depends on estrogen levels.


Subject(s)
Blood Glucose/drug effects , Contraceptives, Oral, Hormonal/pharmacology , Diabetes Mellitus/drug therapy , Ethinyl Estradiol/pharmacology , Hyperprolactinemia/drug therapy , Hypoglycemic Agents/pharmacology , Lactotrophs/drug effects , Metformin/pharmacology , Prolactin/drug effects , Adult , Case-Control Studies , Diabetes Mellitus/blood , Female , Humans , Hyperprolactinemia/blood , Hypoglycemic Agents/administration & dosage , Luteinizing Hormone/blood , Luteinizing Hormone/drug effects , Metformin/administration & dosage , Prediabetic State/blood , Prediabetic State/drug therapy , Prolactin/blood , Young Adult
5.
Front Endocrinol (Lausanne) ; 12: 790441, 2021.
Article in English | MEDLINE | ID: mdl-35058881

ABSTRACT

The role of calcium, but not of other intracellular signaling molecules, in the release of pituitary hormones by exocytosis is well established. Here, we analyzed the contribution of phosphatidylinositol kinases (PIKs) to calcium-driven prolactin (PRL) release in pituitary lactotrophs: PI4Ks - which control PI4P production, PIP5Ks - which synthesize PI(4, 5)P2 by phosphorylating the D-5 position of the inositol ring of PI4P, and PI3KCs - which phosphorylate PI(4, 5)P2 to generate PI(3, 4, 5)P3. We used common and PIK-specific inhibitors to evaluate the strength of calcium-secretion coupling in rat lactotrophs. Gene expression was analyzed by single-cell RNA sequencing and qRT-PCR analysis; intracellular and released hormones were assessed by radioimmunoassay and ELISA; and single-cell calcium signaling was recorded by Fura 2 imaging. Single-cell RNA sequencing revealed the expression of Pi4ka, Pi4kb, Pi4k2a, Pi4k2b, Pip5k1a, Pip5k1c, and Pik3ca, as well as Pikfyve and Pip4k2c, in lactotrophs. Wortmannin, a PI3K and PI4K inhibitor, but not LY294002, a PI3K inhibitor, blocked spontaneous action potential driven PRL release with a half-time of ~20 min when applied in 10 µM concentration, leading to accumulation of intracellular PRL content. Wortmannin also inhibited increase in PRL release by high potassium, the calcium channel agonist Bay K8644, and calcium mobilizing thyrotropin-releasing hormone without affecting accompanying calcium signaling. GSK-A1, a specific inhibitor of PI4KA, also inhibited calcium-driven PRL secretion without affecting calcium signaling and Prl expression. In contrast, PIK93, a specific inhibitor of PI4KB, and ISA2011B and UNC3230, specific inhibitors of PIP5K1A and PIP5K1C, respectively, did not affect PRL release. These experiments revealed a key role of PI4KA in calcium-secretion coupling in pituitary lactotrophs downstream of voltage-gated and PI(4, 5)P2-dependent calcium signaling.


Subject(s)
Calcium/metabolism , Lactotrophs/metabolism , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prolactin/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Calcium Signaling , Exocytosis , Lactotrophs/drug effects , Minor Histocompatibility Antigens/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prolactin/biosynthesis , Prolactin/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Sequence Analysis, RNA , Single-Cell Analysis , Wortmannin/pharmacology
6.
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
7.
Poult Sci ; 99(5): 2708-2717, 2020 May.
Article in English | MEDLINE | ID: mdl-32359608

ABSTRACT

Brooding behavior, a common characteristic of native breeds of the domestic chicken, is marked by elevated prolactin (PRL) levels, which is necessary for incubation and connected with changes in hypothalamic-pituitary-gonadal axis activity. Evidence indicates the serotoninergic system is a potent modulator of PRL secretion. The objective of this study is to investigate whether blocking serotonin synthesis with parachlorophenylalanine (PCPA) prevents incubation behavior in native Polish crested chickens. In addition, we examined the effect of PCPA on the gene expression of the gonadal and lactotrophic axes. Birds were stimulated to broodiness by artificial eggs in nests. At 34 wk of age (April: spring period), the hens were divided into 2 groups (14 hens in each group): control and PCPA-treated (50 mg/kg BW) group. After 5 wk of treatment, the artificial eggs were removed from the nests. Egg production, incubation activity, and levels of plasma ovarian steroids progesterone (P4), testosterone (T), estradiol (E2), and PRL were examined. At the end of the experiment (45 wk of age, June: summer period), ovarian characteristics and mRNA gene expression of gonadal (gonadotropin-releasing hormone [GnRH] I, luteinizing hormone [LH] ß, follicle-stimulating hormone [FSH] ß) and lactotrophic (vasoactive intestinal peptide [VIP], PRL) axes were measured by quantitative real-time PCR. Incubation activity was observed in the hens of both groups but with lower frequency in PCPA-treated birds. Moreover, the PCPA group had a higher cumulative egg production than the controls. During the first six and 8 wk of the experiment, levels of P4 and E2, respectively, were similar in both groups, but all concentrations increased in the PCPA-treated hens after this period. In addition, increased GnRH-I, LHß, and FSHß and decreased VIP mRNA expression was observed in the PCPA group compared with the controls. There were no differences in PRL mRNA expression, the PRL level, and ovarian morphometry between the 2 groups. These results indicate that blockage of serotonin synthesis by PCPA does not effectively prevent incubation in native Polish crested chickens. However, treatment with PCPA increased gonadal axis activity and improved reproductive performance.


Subject(s)
Chickens/physiology , Fenclonine/pharmacology , Lactotrophs/drug effects , Nesting Behavior/drug effects , Ovary/drug effects , Serotonin Antagonists/pharmacology , Animals , Female , Lactotrophs/physiology , Ovary/physiology , Poland , Serotonin/metabolism
8.
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
9.
Am J Physiol Endocrinol Metab ; 316(6): E1036-E1049, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30888862

ABSTRACT

Circulating myostatin-attenuating agents are being developed to treat muscle-wasting disease despite their potential to produce serious off-target effects, as myostatin/activin receptors are widely distributed among many nonmuscle tissues. Our studies suggest that the myokine not only inhibits striated muscle growth but also regulates pituitary development and growth hormone (GH) action in the liver. Using a novel myostatin-null label-retaining model (Jekyll mice), we determined that the heterogeneous pool of pituitary stem, transit-amplifying, and progenitor cells in Jekyll mice depletes more rapidly after birth than the pool in wild-type mice. This correlated with increased levels of GH, prolactin, and the cells that secrete these hormones, somatotropes and lactotropes, respectively, in Jekyll pituitaries. Recombinant myostatin also stimulated GH release and gene expression in pituitary cell cultures although inhibiting prolactin release. In primary hepatocytes, recombinant myostatin blocked GH-stimulated expression of two key mediators of growth, insulin-like growth factor (IGF)1 and the acid labile subunit and increased expression of an inhibitor, IGF-binding protein-1. The significance of these findings was demonstrated by smaller muscle fiber size in a model lacking myostatin and liver IGF1 expression (LID-o-Mighty mice) compared with that in myostatin-null (Mighty) mice. These data together suggest that myostatin may regulate pituitary development and function and that its inhibitory actions in muscle may be partly mediated by attenuating GH action in the liver. They also suggest that circulating pharmacological inhibitors of myostatin could produce unintended consequences in these and possibly other tissues.


Subject(s)
Growth Hormone/metabolism , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Lactotrophs/metabolism , Myostatin/genetics , Pituitary Gland/growth & development , Prolactin/metabolism , Somatotrophs/metabolism , Animals , Cachexia , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Drug Development , Glycoproteins/drug effects , Glycoproteins/metabolism , Growth Hormone/drug effects , Hepatocytes/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 1/drug effects , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Like Growth Factor I/drug effects , Lactotrophs/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Models, Animal , Myostatin/pharmacology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Primary Cell Culture , Prolactin/drug effects , Recombinant Proteins , Somatotrophs/drug effects , Stem Cells
10.
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
11.
Histochem Cell Biol ; 151(4): 291-303, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30511269

ABSTRACT

Diethylstilbestrol (DES), an estrogen agonist, increases prolactin (PRL) cells through transdifferentiation of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) cells to PRL cells as well as proliferation of PRL cells in adult male mouse pituitary. Since hyperacetylation of histone H3 is implicated in the regulation of activation of various genes, we examined the effect of DES on the state of histone H3 acetylation. DES significantly reduced the immunohistochemical signal for acetylated histone H3 at lysine 9 (H3K9ac) in PRL, LH and FSH cells, but not for H3K18ac or H3K23ac. DES-treated mice were injected intraperitoneally with HDAC inhibitors (HDACi), sodium phenylbutyrate (NaPB) or valproic acid (VPA), to mimic the acetylation level of histone H3. As expected, HDACi treatment restored the level of H3K9ac expression in these cells, and also inhibited DES-induced increase in PRL cells. Furthermore, NaPB and VPA also abrogated the effects of DES on the population density of both LH and FSH cells. Similarly, the numbers of proliferating and apoptotic cells in the pituitary in NaPB- or VPA-treated mice were comparable to those of the control mice. Considered together, these results indicated that the acetylation level of histone H3 plays an important role in DES-induced transdifferentiation of LH to PRL cells as well as proliferation of PRL cells.


Subject(s)
Cell Transdifferentiation/drug effects , Gonadotrophs/drug effects , Histone Deacetylase Inhibitors/pharmacology , Lactotrophs/drug effects , Phenylbutyrates/pharmacology , Pituitary Gland/drug effects , Valproic Acid/pharmacology , Acetylation/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Diethylstilbestrol/administration & dosage , Diethylstilbestrol/pharmacology , Gonadotrophs/cytology , Histone Deacetylase Inhibitors/administration & dosage , Histones/analysis , Histones/biosynthesis , Injections, Intraperitoneal , Lactotrophs/cytology , Male , Mice , Mice, Inbred ICR , Phenylbutyrates/administration & dosage , Pituitary Gland/metabolism , Rabbits , Valproic Acid/administration & dosage
12.
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
13.
Neuroendocrinology ; 106(4): 324-334, 2018.
Article in English | MEDLINE | ID: mdl-28848175

ABSTRACT

BACKGROUND/AIMS: During late pregnancy, the blockade of progesterone action by mifepristone (Mp) treatment induces a dopaminergic tone fall that enables naloxone (NAL) administration to release pituitary prolactin (PRL). We determined whether oxytocin (OT), which stimulates PRL secretion acting directly on anterior pituitary lactotrophs, mediates the stimulatory action of Mp and NAL on PRL secretion during late pregnancy. METHODS: On day 19 of pregnancy, circulating and pituitary OT and PRL levels were measured by radioimmunoassay, 10, 20, and 30 min after NAL (given at 17:30 h) in rats pretreated with Mp (at 08:00 h). Pituitary OT receptor (OTR) expression in Mp-treated rats was evaluated by RT-PCR. Activation of OT neurons in Mp-NAL-treated rats was measured counting double immunoreactive neurons for Fos and OT (Fos-OT-ir) in supraoptic nuclei (SON), and medial (PaMM) and lateral magnocellular divisions of paraventricular nuclei. RESULTS: Elevated serum OT and decreased pituitary OT were observed 10 min after NAL administration in both vehicle- and Mp-treated rats. This PRL increase was prevented by previous i.p. administration of an OTR antagonist, but intracerebroventricular OT administration was ineffective. Mp increased pituitary OTR expression at 18:00 h. Only Mp-NAL increased Fos-OT-ir neurons in the PaMM and SON. CONCLUSIONS: These findings suggest that PRL secretion induced by Mp-NAL treatment is preceded by OT release. These results, together with the activation of hypothalamic OT neurons and the higher expression of pituitary OTR, support the hypothesis that, during late pregnancy, OT may act at the pituitary level to facilitate PRL secretion if the inhibitory action of progesterone is blocked.


Subject(s)
Lactotrophs/metabolism , Oxytocin/metabolism , Pregnancy/metabolism , Prolactin/metabolism , Animals , Bodily Secretions , Female , Hormone Antagonists/pharmacology , Lactotrophs/drug effects , Mifepristone/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Rats , Rats, Wistar
14.
Exp Clin Endocrinol Diabetes ; 126(6): 342-348, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29169197

ABSTRACT

BACKGROUND: Metformin was found to reduce circulating levels of pituitary hormones. OBJECTIVE: The purpose of this study was to assess whether sex determines the effect of metformin on lactotroph secretory function. METHODS: The study population included 25 women and 12 men with mildly elevated serum prolactin levels (25-75 ng/mL). Because of concomitant type 2 diabetes, all participants were treated with metformin (3 g daily). Plasma levels of glucose and lipids, HOMA1-IR, serum levels of prolactin, thyrotropin and free thyroid hormones, as well as Jostel's, SPINA-GT and SPINA-GD indices were assessed at baseline and at the end of metformin treatment. RESULTS: The study completed 24 women and 11 men. At baseline, there were no significant differences in circulating levels of glucose and lipids, insulin sensitivity, hormones, Jostel's, SPINA-GT and SPINA-GD indices between women and men. In both men and women, metformin reduced fasting glucose levels and HOMA1-IR. However, only in women metformin decreased elevated prolactin levels and this effect correlated with an improvement insulin sensitivity, as well as with the impact on SPINA-GT. CONCLUSIONS: The results of the study suggest that the effect of metformin on serum prolactin levels is sex-dependent.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hyperprolactinemia/drug therapy , Metformin/therapeutic use , Prolactin/blood , Adolescent , Adult , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Female , Humans , Hyperprolactinemia/blood , Hyperprolactinemia/complications , Lactotrophs/drug effects , Lactotrophs/metabolism , Male , Middle Aged , Pilot Projects , Sex Characteristics , Treatment Outcome , Young Adult
15.
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
16.
J Endocrinol ; 233(1): 37-51, 2017 04.
Article in English | MEDLINE | ID: mdl-28130410

ABSTRACT

In mammals, pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic hormone with diverse functions but its role in prolactin (PRL) regulation is highly controversial. To shed light on Prl regulation by PACAP in fish model, grass carp pituitary cells was used as a model to examine the receptor specificity and signal transduction for PACAP modulation of prl gene expression in the carp pituitary. Using RT-PCR, PACAP-selective PAC1 receptor was detected in carp lactotrophs. In carp pituitary cells, nanomolar doses of PACAP, but not VIP, could elevate Prl secretion and protein production with concurrent rise in prl mRNA and these stimulatory effects were blocked by PACAP antagonist but not VIP antagonist. PACAP-induced prl mRNA expression could be mimicked by activating adenylate cyclase (AC), increasing cAMP level by cAMP analog, or increasing intracellular Ca2+ ([Ca2+]i) by Ca2+ ionophore/voltage-sensitive Ca2+ channel (VSCC) activator. PACAP-induced prl gene expression, however, was attenuated/abolished by suppressing cAMP production, inhibiting PKA activity, blocking [Ca2+]i mobilization and VSCC activation, calmodulin (CaM) antagonism, and inactivation of JNK and CaM Kinase II (CaMK-II). Similar sensitivity to CaM, JNK, and CaMK-II blockade was also noted by substituting cAMP analog for PACAP as the stimulant for prl mRNA expression. These results, as a whole, provide evidence for the first time that (i) PACAP activation of PAC1 receptor expressed in carp lactotrophs could induce Prl synthesis and secretion, and (ii) Prl production induced by PACAP was mediated by upregulation of prl gene expression, presumably via functional coupling of cAMP/PKA-, Ca2+/CaM-, and MAPK-dependent cascades.


Subject(s)
Gene Expression Regulation/drug effects , Lactotrophs/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Gland/drug effects , Prolactin/metabolism , Animals , Calcium/metabolism , Carps , Cyclic AMP/metabolism , Lactotrophs/metabolism , Pituitary Gland/metabolism , Prolactin/genetics , Vasoactive Intestinal Peptide/pharmacology
17.
Naunyn Schmiedebergs Arch Pharmacol ; 390(1): 95-104, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27747371

ABSTRACT

Estrogens facilitate prolactin (PRL) secretion acting on pituitary cells. In GH3 cells, estradiol induces acute action potentials and oscillations of intracellular Ca2+ associated with the secretagogue function. Estradiol modulates several ion channels which may affect the action potential rate and the release of PRL in lactotroph cells, which might depend on its concentration. The aims were to characterize the acute effect of supraphysiological concentrations of estradiol on Ca2+ and noninactivating K+ currents and measure the effect on the spontaneous action potentials and PRL release in the somatolactotroph cell line, GH3. Electrophysiological studies were carried out by voltage- and current-clamp techniques and ELISA determination of PRL secretion. Pharmacological concentrations of estradiol (above 1 µM), without a latency period, blocked Ca2+ channels and noninactivating K+ currents, including the large-conductance voltage- and Ca2+-activated K+ channels (BK), studied in whole-cell nystatin perforated and in excided inside-out patches of GH3 and CHO cells, transiently transfected with the human α-pore forming subunit of BK. The effect on BK was contrary to the agonist effect associated with the regulatory ß1-subunits of the BK, which GH3 cells lack, but its transient transfection did not modify the noninactivating current blockade, suggesting a different mechanism of regulation. Estradiol, at the same concentration range, acutely decreased the frequency of action potentials, an expected effect as consequence of the Ca2+ channel blockade. Despite this, PRL secretion initially increased, followed by a decrease in long-term incubations. This suggests that, in GH3 cells, supraphysiological concentrations of estradiol modulating PRL secretion are partially independent of extracellular Ca2+ influx.


Subject(s)
Action Potentials/drug effects , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Estradiol/pharmacology , Lactotrophs/drug effects , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Prolactin/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Lactotrophs/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Patch-Clamp Techniques , Rats , Time Factors , Transfection
18.
Endocr J ; 64(1): 103-115, 2017 Jan 30.
Article in English | MEDLINE | ID: mdl-27760898

ABSTRACT

Estrogen binds to nuclear estrogen receptors (ERs) to modulate transcription of target genes in estrogen-responsive cells. However, recent studies have shown that estrogen also binds to cytoplasmic membrane ERs to modulate protein kinase signaling cascades, leading to non-genomic actions. We investigated whether either nuclear or membrane ERs, including G protein-coupled estrogen receptor 1 (Gper1), mediate the inhibitory action of estrogen on insulin-like growth factor-1 (IGF-1)-induced proliferation of pituitary lactotrophs in primary culture. The cytoplasmic membrane-impermeable bovine serum albumin-conjugated estradiol (BSA-E2) at 1 nM, an equimolar concentration at which 17ß-estradiol (E2) exerts anti-proliferative effects, did not inhibit IGF-1-induced lactotroph proliferation. In contrast, diethylstilbestrol, which is known to selectively activate nuclear ERs but not membrane ERs, inhibited IGF-1-induced proliferation and modulated mRNA expression of estrogen-responsive genes to a similar degree as E2. Activation of Gper1 by its agonist G-1 inhibited IGF-1-induced proliferation in a dose-dependent manner, but it had little effect on modulation of mRNA expression of estrogen-responsive genes. However, blockade of Gper1 by its antagonist G-15 did not affect the inhibitory action of E2 on IGF-1-induced proliferation. Here, we demonstrate that E2 inhibition of lactotroph proliferation is due to nuclear ER-mediated genomic action. Our results suggest that activation of Gper1 mimics, but does not mediate, the anti-proliferative action of E2 on lactotrophs.


Subject(s)
Cell Proliferation/drug effects , Estradiol/pharmacology , Lactotrophs/drug effects , Receptors, G-Protein-Coupled/agonists , Serum Albumin, Bovine/pharmacology , Animals , Benzodioxoles/pharmacology , Cells, Cultured , Estrogens/pharmacology , Female , Insulin-Like Growth Factor I/pharmacology , Lactotrophs/physiology , Primary Cell Culture , Quinolines/pharmacology , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
19.
J Neuroendocrinol ; 28(12)2016 12.
Article in English | MEDLINE | ID: mdl-27801962

ABSTRACT

Corticotrophin-releasing factor (CRF) regulates the hypothalamic-pituitary-adrenal axis response to stress through its type 1 receptor (CRF1 ) in the corticotrophs of the anterior pituitary. Although CRF1 mRNA expression has been confirmed in the rat pituitary, the distribution pattern of CRF1 protein in the pituitary has not been reported. Therefore, we generated an antiserum against the amino acid fragment corresponding to the 177-188 sequence of the first extracellular loop of the rat CRF1 . Using the antiserum, CRF1 -like immunoreactivity (CRF1 -LI) was detected in the anterior lobe cells of the rat pituitary where some of them expressed intense signals. CRF1 -LI also appeared in the intermediate lobe cells and on the fibre-like elements of the posterior lobe of the pituitary. Dual immunofluorescence labelling showed that corticotrophs exhibited the highest percentage of CRF1 (male: 27.1 ± 3.0%, female: 18.0 ± 3.0%), followed by lactotrophs (male: 6.7 ± 3.0%, female: 12.1 ± 1.3%), gonadotrophs (male: 2.6 ± 1.0%, female: 7.5 ± 0.5%), thyrotrophs (male: 2.9 ± 0.1%, female: 5.3 ± 1.2%) and somatotrophs (male: 1.1 ± 0.3%, female: 1.2 ± 0.5%). The percentage of CRF1 -LI-positive cells that were corticotrophs was significantly higher in male rats than in female rats, whereas CRF1 -LI-positive lactotrophs and gonadotrophs were significantly higher in female rats than in male rats. Almost all of the melanotrophs were positive for CRF1 in the intermediate lobe (98.9 ± 0.2%). CRF1 -LI and the percentage of CRF1 -LI in corticotrophs were decreased in the anterior pituitary, and the distribution patterns were altered from a diffuse to punctate one by adrenalectomy; the changes were restored by treatment with dexamethasone (100 µg/kg bw). These results suggest that CRF1 is involved in the modulation of the functions of the pituitary; moreover, protein expression and the distribution patterns of CRF1 are regulated by glucocorticoids in the rat anterior pituitary.


Subject(s)
Pituitary Gland, Anterior/metabolism , Pituitary Gland/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Adrenalectomy , Animals , Corticotrophs/drug effects , Corticotrophs/metabolism , Dexamethasone/administration & dosage , Female , Glucocorticoids/administration & dosage , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Immunohistochemistry , Lactotrophs/drug effects , Lactotrophs/metabolism , Male , Pituitary Gland/drug effects , Pituitary Gland, Anterior/drug effects , Primary Cell Culture , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/immunology , Somatotrophs/drug effects , Somatotrophs/metabolism , Thyrotrophs/drug effects , Thyrotrophs/metabolism
20.
J Neurosci ; 36(28): 7392-406, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27413150

ABSTRACT

UNLABELLED: Selective serotonin reuptake inhibitors (SSRIs) are commonly prescribed for depression, but sexual side effects often compromise compliance. These reproductive dysfunctions are likely mediated by elevations of the hormone prolactin. Yet, how serotonin (5-HT) and SSRIs cause changes in prolactin secretion is not known. Here, using in vitro whole-cell patch-clamp recordings, we show that 5-HT hyperpolarizes and abolishes phasic discharge in rat neuroendocrine tuberoinfundibular dopamine (TIDA) neurons, the main inhibitor of prolactin secretion. This process is underpinned by 5-HT1A receptor-mediated activation of G-protein-coupled inwardly rectifying K(+)-like currents. We further demonstrate that the SSRIs, fluoxetine and sertraline, directly suppress TIDA neuron activity through parallel effects, independent of 5-HT transmission. This inhibition involves decreased intrinsic excitability and a slowing of TIDA network rhythms. These findings indicate that SSRIs may inhibit neuroendocrine dopamine release through both 5-HT-dependent and -independent actions, providing a mechanistic explanation for, and potential molecular targets for the amelioration of, the hyperprolactinemia and sexual dysfunction associated with these drugs. SIGNIFICANCE STATEMENT: Depression affects approximately one-tenth of the population and is commonly treated with selective serotonin reuptake inhibitors (SSRIs; e.g., Prozac). Yet, many patients withdraw from SSRI therapy due to sexual side effects (e.g., infertility, menstrual disturbances, and impotence). Although it is generally accepted that sexual side effects are due to the ability of these drugs to elevate blood levels of the hormone prolactin, the mechanism for this hormonal imbalance is not known. Here, we show that SSRIs can inhibit hypothalamic dopamine neurons that normally suppress the secretion of prolactin. Intriguingly this inhibition can be explained both by increased serotonin activity and also by parallel serotonin-independent actions.


Subject(s)
Antidepressive Agents/pharmacology , Arcuate Nucleus of Hypothalamus/cytology , Dopaminergic Neurons/drug effects , Lactotrophs/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/pharmacology , Action Potentials/drug effects , Animals , Electric Stimulation , Excitatory Amino Acid Agents/pharmacology , In Vitro Techniques , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/metabolism , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Sodium Channel Blockers/pharmacology , Tetrahydronaphthalenes/pharmacology , Tetrodotoxin/pharmacology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...