Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 260(1)2024 01 01.
Article in English | MEDLINE | ID: mdl-37855268

ABSTRACT

Due to the current limited knowledge about the role of filamin A (FLNA) in pituitary tumour progression, we aimed to analyse FLNA expression levels and its impact on aggressive markers of pituitary neuroendocrine tumours (PitNETs), using an integrative approach of in vivo and in vitro models and human samples. An increase in the expression levels of FLNA was observed in the advanced tumoural stages of the hyperplastic adenomatous pituitary model, concomitant with a decrease in cell proliferation and with a modification in the subcellular localisation of this protein. Similarly, overexpression of FLNA in the somatolactotropic GH3 cell line induced a decrease in the cell proliferation, promoted a migratory phenotype, increased invasion activity, and decreased the prolactin secretion. Cyclin D1 (CCND1) and cyclin-dependent kinase 4 (CDK4) expression increased in both models in correlation with the increase observed in FLNA levels. When human tissues were analysed a significant increase of FLNA was observed in PitNETs compared to normal pituitary gland, with heterogeneous intracellular localisation. Higher levels of FLNA expression were observed in tumours with invasive characteristics. These results underline the crucial roles of FLNA as a modulator of pathological markers and as a potential prognostic marker in pituitary tumours.


Subject(s)
Adenoma , Neuroendocrine Tumors , Pituitary Neoplasms , Humans , Pituitary Neoplasms/metabolism , Filamins/genetics , Filamins/metabolism , Pituitary Gland/metabolism
2.
J Endocr Soc ; 8(1): bvad146, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38045876

ABSTRACT

Serum prolactin (PRL) levels exhibit a gradual rise both in male and female rats from birth to adulthood, with females consistently displaying higher levels compared to age-matched males. This pattern has traditionally been attributed to the development and maturation of endocrine and neuroendocrine networks responsible for regulating PRL synthesis and secretion. However, the effect of dopamine (DA), which acts as an inhibitory factor on lactotroph function, also increases from birth to puberty, particularly in females. Nonetheless, the secretion of PRL remains higher in females compared to males. On the other hand, the observed sex differences in serum PRL levels during early postnatal development cannot be attributed to the influence of estradiol (E2). While serum E2 levels gradually increase after birth, only after 45 days of life do the disparities in E2 levels between females and males become evident. These observations collectively suggest that neither the maturation of hypothalamic DA regulation nor the rise in E2 levels can account for the progressive and sustained elevation in serum PRL levels and the observed sexual dimorphism during postnatal development. This review highlights the importance of recent discoveries in animal models that shed light on inhibitory mechanisms in the control of PRL secretion within the pituitary gland itself, that is intrapituitary mechanisms, with a specific emphasis on the role of transforming growth factor ß1 and activins in PRL secretion.

3.
J Endocrinol ; 258(3)2023 09 01.
Article in English | MEDLINE | ID: mdl-37399522

ABSTRACT

Serum prolactin increases from birth to adulthood in rats, being higher in females from birth. The maturation of hypothalamic/gonadal prolactin-releasing and -inhibiting factors does not explain some sex differences observed. During the first weeks of life, prolactin secretion increases, even when lactotrophs are isolated in vitro, in the absence of those controls, suggesting the participation of intra-pituitary factors in this control. The present work aimed to study the involvement of pituitary activins in the regulation of prolactin secretion during post-natal development. Sex differences were also highlighted. Female and male Sprague-Dawley rats at 11, 23 and 45postnatal days were used. Pituitary expression of activin subunits and activin receptors was maximum in p11 female pituitaries, being even higher than that observed in males. Those expressions decrease with age in females, and then the gender differences disappear at p23. Inhbb expression strongly increases at p45 in males, being the predominant subunit in this sex in adulthood. Activin inhibition of prolactin is mediated by the inhibition of Pit-1 expression. This action involves not only the canonical pSMAD pathway but also the phosphorylation of p38MAPK. At p11, almost all lactotrophs express p-p38MAPK in females, and its expression decreases with age with a concomitant increase in Pit-1. Our findings suggest that the inhibitory regulation of pituitary activins on prolactin secretion is sex specific; this regulation is more relevant in females during the first week of life and decreases with age; this intra-pituitary regulation is involved in the sex differences observed in serum prolactin levels during postnatal development.


Subject(s)
Lactotrophs , Prolactin , Female , Rats , Male , Animals , Prolactin/metabolism , Activins/metabolism , Rats, Sprague-Dawley , Pituitary Gland/metabolism , Lactotrophs/metabolism , Transcription Factors/metabolism
5.
Endocrinology ; 163(9)2022 09 01.
Article in English | MEDLINE | ID: mdl-35863039

ABSTRACT

Sex differences in the control of prolactin secretion are well documented. Sex-related differences in intrapituitary factors regulating lactotroph function have recently attracted attention. Sex differences in prolactinoma development are well documented in clinic, prolactinomas being more frequent in women but more aggressive in men, for poorly understood reasons. Kallikrein, the enzyme releasing kinins has been found in the pituitary, but there is no information on pituitary kinin receptors and their function. In the present work, we characterized pituitary bradykinin receptors (BRs) at the messenger RNA and protein levels in 2 mouse models of prolactinoma, Drd2 receptor gene inactivation and hCGß gene overexpression, in both males and females, wild type or genomically altered. BR B2 (B2R) accounted for 97% or more of total pituitary BRs in both models, regardless of genotype, and was present in lactotrophs, somatotrophs, and gonadotrophs. Male pituitaries displayed higher level of B2R than females, regardless of genotype. Pituitary B2R gene expression was downregulated by estrogen in both males and females but only in females by dopamine. Activation of B1R or B2R by selective pharmacological agonists induced prolactin release in male pituitaries but inhibited prolactin secretion in female pituitaries. Increased B2R content was observed in pituitaries of mutated animals developing prolactinomas, compared to their respective wild-type controls. The present study documents a novel sex-related difference in the control of prolactin secretion and suggests that kinins are involved, through B2R activation, in lactotroph function and prolactinoma development.


Subject(s)
Pituitary Neoplasms , Prolactinoma , Animals , Female , Humans , Kinins , Male , Mice , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Prolactin/metabolism , Prolactinoma/genetics , Prolactinoma/metabolism , Receptor, Bradykinin B2/agonists , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/metabolism , Receptors, Bradykinin
6.
Endocr Relat Cancer ; 29(6): 359-373, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35324456

ABSTRACT

Among pituitary adenomas, prolactinomas are the most frequently diagnosed (about 50%). Dopamine agonists are generally effective in the treatment of prolactinomas. However, a subset of about 25% of patients does not respond to these agents. The management of drug-resistant prolactinomas remains a challenge for endocrinologists and new inhibitory treatments are needed. Pituitary activins inhibit lactotroph function. Its expression and action were found reduced in animal models of lactotroph hyperplasia (female mice overexpressing the B subunit of the human chorionic gonadotrophin and female mice knockout for dopamine receptor type 2). In these models, an oophorectomy avoids prolactinoma development. Hormonal replacement with oestradiol and/or progesterone is not enough to reach the tumor size observed in transgenic females. We postulated that the loss of gonadal inhibins after an oophorectomy contributes to prevent hyperplasia development. Here, we demonstrated that an oophorectomy at 2 months age recovers the following in adulthood: (i) pituitary activin expression, (ii) activin receptor expression specifically in lactotroph population, (iii) activin biological activity in lactotrophs with a concomitant reduction of Pit-1 expression. To summarize, when an oophorectomy is performed, inhibins are lost and the inhibitory action of pituitary activins on lactotroph population is recovered, helping to prevent lactotroph hyperplasia development. These results emphasize the importance of the inhibitory action of activins on lactotroph function, positioning activins as a good therapeutic target for the treatment of resistant prolactinomas.


Subject(s)
Lactotrophs , Pituitary Neoplasms , Prolactinoma , Activins/metabolism , Adult , Animals , Female , Humans , Hyperplasia , Inhibins/metabolism , Inhibins/therapeutic use , Lactotrophs/metabolism , Lactotrophs/pathology , Mice , Ovariectomy , Pituitary Neoplasms/metabolism , Prolactin/metabolism , Prolactinoma/metabolism , Prolactinoma/prevention & control
7.
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
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.
Steroids ; 152: 108496, 2019 12.
Article in English | MEDLINE | ID: mdl-31521709

ABSTRACT

Progesterone (P4) has controversial physiological effects on the regulation of the lactotroph population. While some studies have shown a negative role for P4 in prolactin secretion and lactotroph proliferation, antagonizing estradiol effects, others demonstrated a proliferative role of P4 at the pituitary level. Usually, progesterone actions in the pituitary gland were studied through their classical, genomic pathways triggered by nuclear progesterone receptors (nPRs). However, in 2003, the scene became more complex with the discovery of another group of progesterone receptors involved in rapid, non-genomic P4 effects: the membrane progesterone receptors (mPRs), which are members of the progesterone and adipoQ receptor (PAQR) family. This review examines the historical background and current data on the study of progesterone actions on PRL secretion providing new evidence of P4 effects at the hypothalamic and at the pituitary level through non-classic P4-receptors. In addition, we explore the role of progesterone in the development of experimental prolactinomas, a controversial topic in the literature.


Subject(s)
Pituitary Neoplasms/metabolism , Progesterone/metabolism , Prolactin/metabolism , Prolactinoma/metabolism , Animals , Humans
10.
Endocr Relat Cancer ; 26(5): 497-510, 2019 05.
Article in English | MEDLINE | ID: mdl-30856609

ABSTRACT

Membrane progesterone receptors are known to mediate rapid nongenomic progesterone effects in different cell types. Recent evidence revealed that mPRα is highly expressed in the rat pituitary, being primarily localized in lactotrophs, acting as an intermediary of P4-inhibitory actions on prolactin secretion. The role of mPRs in prolactinoma development remains unclear. We hypothesize that mPR agonists represent a novel tool for hyperprolactinemia treatment. To this end, pituitary expression of mPRs was studied in three animal models of prolactinoma. Expression of mPRs and nuclear receptor was significantly decreased in tumoral pituitaries compared to normal ones. However, the relative proportion of mPRα and mPRß was highly increased in prolactinomas. Interestingly, the selective mPR agonist (Org OD 02-0) significantly inhibited PRL release in both normal and tumoral pituitary explants, displaying a more pronounced effect in tumoral tissues. As P4 also regulates PRL secretion indirectly, by acting on dopaminergic neurons, we studied mPR involvement in this effect. We found that the hypothalamus has a high expression of mPRs. Interestingly, both P4 and OrgOD 02-0 increased dopamine release in hypothalamus explants. Moreover, in an in vivo treatment, that allows both, pituitary and hypothalamus actions, the mPR agonist strongly reduced the hyperprolactinemia in transgenic females carrying prolactinoma. Finally, we also found and interesting gender difference: males express higher levels of pituitary mPRα/ß, a sex that does not develop prolactinoma in these mice models. Taken together, these findings suggest mPRs activation could represent a novel tool for hyperprolactinemic patients, especially those that present resistance to dopaminergic drugs.


Subject(s)
Pituitary Neoplasms/prevention & control , Progesterone/pharmacology , Prolactin/metabolism , Prolactinoma/prevention & control , Receptors, Dopamine D2/physiology , Receptors, Progesterone/agonists , Animals , Chorionic Gonadotropin, beta Subunit, Human/genetics , Chorionic Gonadotropin, beta Subunit, Human/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Pituitary Neoplasms/etiology , Pituitary Neoplasms/pathology , Prolactinoma/etiology , Prolactinoma/pathology , Rats , Signal Transduction
11.
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
12.
Front Neuroendocrinol ; 50: 118-122, 2018 07.
Article in English | MEDLINE | ID: mdl-29074127

ABSTRACT

Prolactinomas are the most frequent functioning pituitary adenomas, and sex differences in tumor size, behavior and incidence have been described. These differences have been associated with earlier diagnosis in woman, as well as with serum estradiol levels. Experimental models of prolactinomas in rodents also show a higher incidence in females, and recent findings suggest that gender differences in the transforming growth factor beta 1 (TGFß1) system might be involved in the sex-specific development of prolactinomas in these models. The aim of this review is to summarize the literature supporting the important role of TGFß1 as a local modulator of pituitary lactotroph function and to provide recent evidence for TGFß1 involvement in the sex differences found in prolactinoma development in animal models.


Subject(s)
Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Sex Characteristics , Transforming Growth Factor beta1/metabolism , Animals , Female , Humans , Male
13.
J Endocrinol ; 232(3): 535-546, 2017 03.
Article in English | MEDLINE | ID: mdl-28096433

ABSTRACT

Female transgenic mice that overexpress the human chorionic gonadotrophin ß subunit (hCGß+) develop prolactinomas, whereas hCGß+ males do not. The high levels of circulating hCG induce massive luteinization in the ovary of hCGß+ females, and progesterone becomes the primary steroid hormone produced, but estradiol remains at physiological level. The involvement of high levels of progesterone in lactotroph proliferation is not clearly understood; hence, the pathogenesis of prolactinomas in hCGß+ females remains unclear. TGFß1 is an inhibitor of lactotroph function, and the reduced TGFß1 activity found in prolactinomas has been proposed to be involved in tumor development. The aim of the present work was to study the role of TGFß1 in the gender-specific development of prolactinomas in hCGß+ mice. We compared the expression of different components of the pituitary TGFß1 system in males and females in this model. We found reduced TGFß1 levels, reduced expression of TGFß1 target genes, TGFß1 receptors, Ltbp1, Smad4 and Smad7 in hCGß+ female pituitaries. However, no differences were found between the transgenic and wild-type male pituitaries. We postulate that decreased pituitary TGFß1 activity in hCGß+ females is involved in the development of prolactinomas. In fact, we demonstrated that an in vivo treatment carried out for increasing pituitary TGFß1 activity, was successful in reducing the prolactinoma development, and the hyperprolactinemia in hCGß+ females. Moreover, the stronger TGFß1 system found in males could protect them from excessive lactotroph proliferation. Sex differences in the regulation of the pituitary TGFß1 system could explain gender differences in the incidence of prolactinoma.


Subject(s)
Chorionic Gonadotropin, beta Subunit, Human/metabolism , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Sex Characteristics , Transforming Growth Factor beta1/metabolism , Animals , Chorionic Gonadotropin, beta Subunit, Human/genetics , Female , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Male , Mice , Mice, Transgenic , Pituitary Gland/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Prolactinoma/genetics , Prolactinoma/pathology , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism
14.
J Endocrinol ; 228(3): R73-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26698564

ABSTRACT

Prolactinomas are the most frequently observed pituitary adenomas and most of them respond well to conventional treatment with dopamine agonists (DAs). However, a subset of prolactinomas fails to respond to such therapies and is considered as DA-resistant prolactinomas (DARPs). New therapeutic approaches are necessary for these tumors. Transforming growth factor ß1 (TGFß1) is a known inhibitor of lactotroph cell proliferation and prolactin secretion, and it partly mediates dopamine inhibitory action. TGFß1 is secreted to the extracellular matrix as an inactive latent complex, and its bioavailability is tightly regulated by different components of the TGFß1 system including latent binding proteins, local activators (thrombospondin-1, matrix metalloproteases, integrins, among others), and TGFß receptors. Pituitary TGFß1 activity and the expression of different components of the TGFß1 system are regulated by dopamine and estradiol. Prolactinomas (animal models and humans) present reduced TGFß1 activity as well as reduced expression of several components of the TGFß1 system. Therefore, restoration of TGFß1 inhibitory activity represents a novel therapeutic approach to bypass dopamine action in DARPs. The aim of this review is to summarize the large literature supporting TGFß1 important role as a local modulator of pituitary lactotroph function and to provide recent evidence of the restoration of TGFß1 activity as an effective treatment in experimental prolactinomas.


Subject(s)
Drug Resistance, Neoplasm , Pituitary Gland/metabolism , Pituitary Neoplasms/drug therapy , Prolactinoma/drug therapy , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/physiology , Animals , Cell Proliferation , Dopamine/physiology , Dopamine Agonists/therapeutic use , Estradiol/physiology , Humans , Lactotrophs/physiology , Pituitary Neoplasms/physiopathology , Prolactin/antagonists & inhibitors , Prolactin/metabolism , Prolactinoma/physiopathology
15.
Endocrinology ; 154(11): 4192-205, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008346

ABSTRACT

Dopamine and estradiol interact in the regulation of lactotroph cell proliferation and prolactin secretion. Ablation of the dopamine D2 receptor gene (Drd2(-/-)) in mice leads to a sexually dimorphic phenotype of hyperprolactinemia and pituitary hyperplasia, which is stronger in females. TGF-ß1 is a known inhibitor of lactotroph proliferation. TGF-ß1 is regulated by dopamine and estradiol, and it is usually down-regulated in prolactinoma experimental models. To understand the role of TGF-ß1 in the gender-specific development of prolactinomas in Drd2(-/-) mice, we compared the expression of different components of the pituitary TGF-ß1 system, including active cytokine content, latent TGF-ß-binding protein isoforms, and possible local TGF-ß1 activators, in males and females in this model. Furthermore, we evaluated the effects of dopamine and estradiol administration to elucidate their role in TGF-ß1 system regulation. The expression of active TGF-ß1, latent TGF-ß-binding protein isoforms, and several putative TGF-ß1 activators evaluated was higher in male than in female mouse pituitary glands. However, Drd2(-/-) female mice were more sensitive to the decrease in active TGF-ß1 content, as reflected by the down-regulation of TGF-ß1 target genes. Estrogen and dopamine caused differential regulation of several components of the TGF-ß1 system. In particular, we found sex- and genotype- dependent regulation of active TGF-ß1 content and a similar expression pattern for 2 of the putative TGF-ß1 activators, thrombospondin-1 and kallikrein-1, suggesting that these proteins could mediate TGF-ß1 activation elicited by dopamine and estradiol. Our results indicate that (1) the loss of dopaminergic tone affects the pituitary TGF-ß1 system more strongly in females than in males, (2) males express higher levels of pituitary TGF-ß1 system components including active cytokine, and (3) estradiol negatively controls most of the components of the system. Because TGF-ß1 inhibits lactotroph proliferation, we propose that the higher levels of the TGF-ß1 system in males could protect or delay the development of prolactinomas in Drd2(-/-) male mice.


Subject(s)
Pituitary Gland/metabolism , Prolactinoma/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Female , Gene Expression Regulation/physiology , Genotype , Integrins/genetics , Integrins/metabolism , Male , Mice , Mice, Knockout , Pituitary Neoplasms/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Sex Factors , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Tissue Kallikreins/genetics , Tissue Kallikreins/metabolism , Transforming Growth Factor beta1/genetics
16.
Endocrinology ; 153(8): 3861-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22700773

ABSTRACT

Prolactinomas are the most prevalent type of secreting pituitary tumors in humans and generally respond well to a medical therapy with dopamine agonists. However, for patients exhibiting resistance to dopaminergic drugs, alternative treatments are desired. Antiangiogenic strategies might represent a potential therapy for these tumors. Thrombospondin 1 (TSP-1) is a large multifunctional glycoprotein involved in multiple biological processes including angiogenesis, apoptosis, and activation of TGF-ß1. Because tumors that overexpress TSP-1 grow more slowly, have fewer metastases, and have decreased angiogenesis, TSP-1 provides a novel target for cancer treatment. ABT-510 and ABT-898 are TSP-1 synthetic analogs that mimic its antiangiogenic action. In the present study, we explored the potential effect of ABT-510 and ABT-898 on experimental prolactinomas induced by chronic diethylstilbestrol (DES) treatment in female rats. We demonstrated that a 2-wk treatment with ABT-510 and ABT-898 counteracted the increase in pituitary size and serum prolactin levels as well as the pituitary proliferation rate induced by DES. These inhibitory effects on tumor growth could be mediated by the antiangiogenic properties of the drugs. We also demonstrated that ABT-510 and ABT-898, in addition to their described antiangiogenic effects, increased active TGF-ß1 level in the tumors. We postulate that the recovery of the local cytokine activation participates in the inhibition of lactotrope function. These results place these synthetic TSP-1 analogs as potential alternative or complementary treatments in dopamine agonist-resistant prolactinomas.


Subject(s)
Oligopeptides/therapeutic use , Prolactinoma/drug therapy , Thrombospondin 1/chemistry , Transforming Growth Factor beta1/metabolism , Animals , Diethylstilbestrol/toxicity , Female , Oligopeptides/chemistry , Prolactinoma/chemically induced , Prolactinoma/metabolism , Rats , Rats, Sprague-Dawley
17.
Endocrinology ; 152(7): 2722-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21521749

ABSTRACT

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


Subject(s)
Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Estradiol/analogs & derivatives , Gene Expression Regulation/drug effects , Pituitary Gland, Anterior/drug effects , Receptors, Dopamine D2/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Proliferation , Cells, Cultured , Dopamine Agonists/therapeutic use , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Hyperprolactinemia/drug therapy , Lactotrophs/drug effects , Lactotrophs/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Prolactin/blood , Prolactin/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics
18.
Neuroendocrinology ; 92(4): 207-14, 2010.
Article in English | MEDLINE | ID: mdl-20975260

ABSTRACT

Dopamine D2 receptor (D2R) participation in prolactin regulation is well documented, but the role of D2Rs in the control of other hormones involved in growth, food intake and glucose metabolism has not been extensively studied. The study of D2R knockout mice (Drd2(-/-)) puts forward new insights into the role of the D2R in growth hormone (GH)-releasing hormone-GH regulation, peptides involved in food intake, glucose homeostasis, as well as in prolactinoma development. The expected phenotype of chronic hyperprolactinemia and prolactinoma development was found in the Drd2(-/-) mouse, and this model constitutes a valuable tool in the study of dopamine-resistant prolactinomas. Unexpectedly, these mice were growth retarded, and the importance of functional hypothalamic D2Rs in the neonatal period was revealed. In the Drd2(-/-) mouse there was a failure of high neonatal GH levels and therefore the expansion of pituitary somatotropes was permanently altered. These mice also had increased food intake, and a sexually dimorphic participation of the D2R in food intake regulation is suggested. The effect described is probably secondary to D2R regulation of prolactin secretion. Furthermore, the negative modulation of D2Rs on α-melanocyte-stimulating hormone release and positive action on the hypothalamic expression of orexins reveals the complex D2R regulation of food intake. Finally, pancreatic D2Rs inhibit glucose-stimulated insulin release. Lack of dopaminergic inhibition throughout development in the Drd2(-/-) mouse may exert a gradual deteriorating effect on insulin homeostasis, so that eventually glucose intolerance develops. These results highlight the complex endocrine actions of the D2Rs at different levels, hypothalamus, pituitary or pancreas, which function to improve fitness, reproductive success and survival.


Subject(s)
Endocrine System/physiology , Metabolism/genetics , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology , Animals , Eating/genetics , Eating/physiology , Endocrine System/metabolism , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/metabolism , Mice , Mice, Knockout , Prolactin/metabolism , Receptors, Dopamine D2/metabolism
19.
Front Horm Res ; 38: 59-69, 2010.
Article in English | MEDLINE | ID: mdl-20616496

ABSTRACT

The role of dopaminergic receptors in the control of GH release remains controversial. The dopamine receptor 2 (D2R) knockout mouse represents a useful model to study the participation of the D2R on growth and GHRH-GH regulation. These knockout mice have hyperprolactinemia and lactotrope hyperplasia, but unexpectedly, they are also growth retarded. In D2R knockout mice there is a significant decrease in somatotrope population, which is paralleled by decreased GH content and output from pituitary cells. The sensitivity of GHRH-induced GH and cAMP release is similar between genotypes, even though the response amplitude is lower in knockouts. We point to an involvement of D2R signaling at the hypothalamic level as dopamine did not release GH acting at the pituitary level, and both somatostatin and GHRH mRNA expression are altered in knockout mice. The similarity of the pituitary defect in the D2R knockout mouse to that of GHRH deficient models suggests a probable mechanism. Loss of dopamine signaling via hypothalamic D2Rs at a critical age may cause inadequate GHRH secretion subsequently leading to inappropriate somatotrope lineage development. Furthermore, GH pulsatility, which depends on a regulated temporal balance between GHRH and somatostatin output might be compromised in D2R knockout mice, leading to lower IGF-I, and growth retardation.


Subject(s)
Dopamine/physiology , Growth Hormone-Releasing Hormone/physiology , Growth Hormone/physiology , Neurotransmitter Agents/physiology , Acromegaly/drug therapy , Animals , Growth , Humans , Mice , Receptors, Dopamine D2/physiology
20.
Am J Physiol Endocrinol Metab ; 293(5): E1341-51, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17848635

ABSTRACT

Dopamine D2 receptor (D2R) knockout (KO) female mice develop chronic hyperprolactinemia and pituitary hyperplasia. Our objective was to study the expression of the mitogen fibroblast growth factor (FGF2) and its receptor, FGFR1, comparatively in pituitaries from KO and wild-type (WT) female mice. We also evaluated FGF2 subcellular localization and FGF2 effects on pituitary function. FGF2-induced prolactin release showed a similar response pattern in both genotypes, even though basal and FGF2-stimulated release was higher in KO. FGF2 stimulated pituitary cellular proliferation (MTS assay and [(3)H]thymidine incorporation), with no differences between genotypes. FGF2 concentration (measured by ELISA) in whole pituitaries or cultured cells was lower in KO (P < 0.00001 and 0.00014). Immunofluorescence histochemistry showed less FGF2 in pituitaries from KO females and revealed a distinct FGF2 localization pattern between genotypes, being predominantly nuclear in KO and cytosolic in WT pituitaries. Finally, FGF2 could not be detected in the conditioned media from pituitary cultures of both genotypes. FGFR1 levels (Western blot and immunohistochemistry) were higher in pituitaries of KO. Basal concentration of phosphorylated ERKs was lower in KO cells (P = 0.018). However, when stimulated with FGF2, a significantly higher increment of ERK phosphorylation was evidenced in KO cells (P < or = 0.02). We conclude that disruption of the D2R caused an overall decrease in pituitary FGF2 levels, with an increased distribution in the nucleus, and increased FGFR1 levels. These results are important in the search for reliable prognostic indicators for patients with pituitary dopamine-resistant prolactinomas, which will make tumor-specific therapy possible.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Prolactinoma/metabolism , Receptors, Dopamine D2/deficiency , Animals , Blotting, Western , Cell Growth Processes/physiology , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hyperplasia , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation , Pituitary Gland, Anterior/cytology , Prolactin/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Dopamine D2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...