Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
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.
Neuroendocrinology ; 113(7): 705-718, 2023.
Article in English | MEDLINE | ID: mdl-36716712

ABSTRACT

INTRODUCTION: Macroautophagy is a lysosome-mediated degradation process that controls the quality of cytoplasmic components and organelles, with its regulation depending on autophagy-related proteins (Atg) and with Beclin1/Atg6 and microtubule-associated protein light chain 3 (LC3/Atg8) being key players in the mammalian autophagy. As reports on this mechanism in the field of pituitary neuropathology and neuroendocrinology are scarce, our study analyzed the ultrastructural signs of macroautophagy and the expression of Beclin1 and LC3 proteins in human functioning PitNETs and in experimental pituitary tumors. METHODS: A group of humans functioning PitNETs and an experimental lactotroph model in rats of the F344 strain stimulated with estradiol benzoate (BE) were used. Ultrastructural and molecular evidence of the macroautophagic process was evaluated using different techniques. RESULTS: In functioning PitNETs cohort, 60% exhibited evidence of macroautophagy, with a significant difference found for Beclin1 and LC3 between macro- and micro-PitNETs (p < 0.05). In the experimental model, the expression of both Beclin1 and LC3 proteins was immunopositive in normal and tumoral glands when analyzed by immunofluorescence, Western blot, and immunohistochemistry. In the experimental model, protein expression was associated with increased glandular size and weight. CONCLUSIONS: Our study revealed evidence of macroautophagy at the pituitary level and the important role of Beclin1 and LC3 in the progression of functioning PitNETs, implying that this mechanism participate in regulating pituitary cell growth.


Subject(s)
Macroautophagy , Pituitary Neoplasms , Humans , Rats , Animals , Beclin-1 , Rats, Inbred F344 , Autophagy , Microtubule-Associated Proteins/metabolism , Mammals/metabolism
5.
Rev Fac Cien Med Univ Nac Cordoba ; 78(4): 423-429, 2021 12 28.
Article in Spanish | MEDLINE | ID: mdl-34962746

ABSTRACT

Pituitary adenomas are intracranial neoplasms that originate from the adeno-pituitary cells, are mostly benign and slow growing. However, a small percentage can be aggressive and spread locally and / or remotely as malignancies. In recent years, progress has been made in understanding the biology of pituitary tumors, identifying mutations in the germline, somatic lines, and epigenetic mechanisms. Objective: review the updated bibliography on the mechanisms of pituitary tumorigenesis. Data source: bibliographic search was performed using the MEDLINE (PubMed), LILACS and Google Scholar databases since 2010 to April 2020. Conclusion: Knowledge and information on pituitary tumorogenesis mechanisms increased in recent decades, and new neoplastic pathways are recognized. However, there are currently few therapeutic approaches to act specifically on the underlying tumor genesis pathway identified in each case.


Los adenomas hipofisarios son neoplasias intracraneales que surgen de las células del lóbulo anterior de la glándula, benignos y de crecimiento lento en su gran mayoría. Sin embargo, un pequeño porcentaje puede mostrar un comportamiento clínicamente agresivo y diseminarse localmente y/o a distancia como verdaderas neoplasias malignas. Durante los últimos años, se observó un importante avance en el conocimiento de la biología de los tumores hipofisarios, identificándose mutaciones de las líneas germinal, somática y mecanismos epigenéticos. Objetivo: revisar la bibliografía actualizada sobre los mecanismos que contribuyen a la tumorogénesis hipofisaria. Fuente de datos: se realizó una búsqueda bibliográfica utilizando las bases de datos de MEDLINE (PubMed), LILACS y Google Scholar desde 2010 hasta abril de 2020. Conclusión: El conocimiento e información sobre los mecanismos asociados a la formación de tumores hipofisarios incrementó a lo largo de las últimas décadas, y se reconocen nuevas vías de desarrollo neoplásico. Sin embargo, en la actualidad existen pocos enfoques terapéuticos para actuar específicamente sobre la vía de génesis tumoral subyacente identificada en cada caso.


Subject(s)
Adenoma , Pituitary Neoplasms , Carcinogenesis/genetics , Humans , Mutation , Pituitary Neoplasms/genetics
6.
J Neuroendocrinol ; 33(12): e13051, 2021 12.
Article in English | MEDLINE | ID: mdl-34708474

ABSTRACT

To investigate the putative stem cell/tumor stem cell (SC/TSC) niche contribution to hyperplasic/adenomatous pituitary lesions, we analyzed variation in the pituitary stem cell population during the development of experimental pituitary tumors. Pituitary tumors were induced in female F344 rats with estradiol benzoate for 5, 10, 20 and 30 days. Cells positive for GFRa2, Sox2, Sox9, Nestin, CD133 and CD44 were identified in the marginal zone and in the adenoparenchyma in both control and 30D groups, with predominant adenoparenchyma localization of GRFa2 and SOX9 found in tumoral pituitaries. GFRa2, Nestin, CD133 and CD44 were upregulated at the initial stages of tumor growth, whereas Sox9 significantly decreased at 5D, with Sox2 remaining invariable during the hyperplasic/adenomatous development. In addition, isolated pituispheres from normal and tumoral pituitary glands enriched in SC/TSC were characterized. Pituispheres from the 30D glands were positive for the above-mentioned markers and showed a significant increase in the proliferation. In conclusion, our data revealed pituitary SC pool fluctuations during hyperplastic/adenomatous development, with differential localization of the SC/TSC niche in this process. These findings may help to provide a better understanding of these cell populations, which is crucial for achieving advancements in the field of pituitary tumor biology.


Subject(s)
Adenoma/pathology , Pituitary Neoplasms/pathology , Stem Cell Niche/physiology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Pituitary Gland/pathology , Pituitary Gland/physiology , Rats , Rats, Inbred F344 , Tumor Microenvironment/physiology
7.
Endocr Relat Cancer ; 25(10): 837-852, 2018 10.
Article in English | MEDLINE | ID: mdl-29875136

ABSTRACT

In pituitary adenomas, early recurrences and resistance to conventional pharmacotherapies are common, but the mechanisms involved are still not understood. The high expression of epidermal growth factor receptor 2 (HER2)/extracellular signal-regulated kinase (ERK1/2) signal observed in human pituitary adenomas, together with the low levels of the antimitogenic transforming growth factor beta receptor 2 (TBR2), encouraged us to evaluate the effect of the specific HER2 inhibition with trastuzumab on experimental pituitary tumor cell growth and its effect on the antiproliferative response to TGFB1. Trastuzumab decreased the pituitary tumor growth as well as the expression of ERK1/2 and the cell cycle regulators CCND1 and CDK4. The HER2/ERK1/2 pathway is an attractive therapeutic target, but its intricate relations with other signaling modulators still need to be unraveled. Thus, we investigated possible cross-talk with TGFB signaling, which has not yet been studied in pituitary tumors. In tumoral GH3 cells, co-incubation with trastuzumab and TGFB1 significantly decreased cell proliferation, an effect accompanied by a reduction in ERK1/2 phosphorylation, an increase of SMAD2/3 activation. In addition, through immunoprecipitation assays, a diminution of SMAD2/3-ERK1/2 and an increase SMAD2/3-TGFBR1 interactions were observed when cells were co-incubated with trastuzumab and TGFB1. These findings indicate that blocking HER2 by trastuzumab inhibited pituitary tumor growth and modulated HER2/ERK1/2 signaling and consequently the anti-mitogenic TGFB1/TBRs/SMADs cascade. The imbalance between HER2 and TGFBRs expression observed in human adenomas and the response to trastuzumab on experimental tumor growth may make the HER2/ERK1/2 pathway an attractive target for future pituitary adenoma therapy.


Subject(s)
Adenoma/metabolism , Cell Proliferation/drug effects , Pituitary Neoplasms/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Trastuzumab/pharmacology , Adenoma/pathology , Adult , Cell Cycle/drug effects , Female , Humans , Male , Middle Aged , Phosphorylation , Pituitary Neoplasms/pathology , Young Adult
8.
J Mol Histol ; 48(3): 209-218, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28374152

ABSTRACT

Uric acid (UA) has been associated with renal fibrosis and progression of chronic kidney disease. However, the underlying mechanisms of this process have still not been identified. Here, we studied the role of the innate imunity receptor NLRP3/ASC in UA induced epithelial-mesenchymal transition (EMT) in kidney. Wistar rats were fed with oxonic acid 2% and UA 2% (OXA + U), OXA + U plus allopurinol (ALL) or regular chow (C) for 7 weeks. We analyzed the presence of EMT markers, the expression of NLRP3, ASC, Caspase-1 and Smad 2/3 molecules and the mitochondrial morphological and functional characteristics. High UA induced renal fibrosis, mild chronic inflammation, as well as morphological and biochemical evidence of EMT. High UA also increased the expression of NLRP3/ASC with activation of both inflammasome related caspase-1 and inflammasome unrelated Smad 2/3 pathways. Ultrastructural co-localization of NLRP3 and Smad 2/3 indicated physical interaction between the two molecules. No morphological or functional changes were found between mitochondria exposed to high UA. In conclusion, kidney epithelial NLRP3/ASC expression was increased in high UA state in rats and both inflammasome related caspase-1 and non-inflammasome related P-Smad 2/3 pathways were associated with the observed EMT, inflammation and fibrosis induced by UA in the kidney.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Inflammasomes/metabolism , Uric Acid/pharmacology , Animals , Caspase 1/metabolism , Fibrosis/chemically induced , Inflammation/chemically induced , Kidney Diseases/metabolism , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats, Wistar , Smad Proteins/metabolism
9.
Mol Cell Biochem ; 390(1-2): 1-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24623265

ABSTRACT

Interest in biochemistry of organoselenium compound has increased in the last decades, mainly due to their chemical and biological activities. Here, we investigated the protective effect of diphenyl diselenide (PhSe)2 (5 µmol/kg), in a mouse model of methylmercury (MeHg)-induced brain toxicity. Swiss male mice were divided into four experimental groups: control, (PhSe)2 (5 µmol/kg, subcutaneous administration), MeHg (40 mg/L, in tap water), and MeHg + (PhSe)2. After the treatment (21 days), the animals were killed and the cerebral cortex was analyzed. Electron microscopy indicated an enlarged and fused mitochondria leading to a reduced number of organelles, in the MeHg-exposed mice. Furthermore, cortical creatine kinase activity, a sensitive mitochondrial oxidative stress sensor, was almost abolished by MeHg. Subcutaneous (PhSe)2 co-treatment rescued from MeHg-induced mitochondrial alterations. (PhSe)2 also behaved as an enhancer of mitochondrial biogenesis, by increasing cortical mitochondria content in mouse-receiving (PhSe)2 alone. Mechanistically, (PhSe)2 (1 µM; 24 h) would trigger the cytoprotective Nrf-2 pathway for activating target genes, since astroglial cells exposed to the chalcogen showed increased content of hemeoxygenase type 1, a sensitive marker of the activation of this via. Thus, it is proposed that the (PhSe)2-neuroprotective effect might be linked to its mitoprotective activity.


Subject(s)
Benzene Derivatives/administration & dosage , Brain/metabolism , Heme Oxygenase-1/biosynthesis , Mitochondria/metabolism , Organoselenium Compounds/administration & dosage , Animals , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Male , Mercury Poisoning, Nervous System/metabolism , Mercury Poisoning, Nervous System/pathology , Methylmercury Compounds/toxicity , Mice , Mitochondria/drug effects , Oxidative Stress/drug effects
10.
Exp Cell Res ; 319(19): 3020-34, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23973924

ABSTRACT

Toll like receptor 4 (TLR4) has been characterized for its ability to recognize bacterial endotoxin lipopolysaccharide (LPS). Considering that infections or inflammatory processes might contribute to the progression of pituitary tumors, we analyzed the TLR4 functional role by evaluating the LPS effect on lactotroph proliferation in primary cultures from experimental pituitary tumors, and examined the involvement of PI3K-Akt and NF-κB activation in this effect. In addition, the role of 17ß-estradiol as a possible modulator of LPS-induced PRL cell proliferation was further investigated. In estrogen-induced hyperplasic pituitaries, LPS triggered lactotroph cell proliferation. However, endotoxin failed to increase the number of lactotrophs taking up BrdU in normal pituitaries. Moreover, incubation with anti-TLR4 antibody significantly reduced LPS-induced lactotroph proliferation, suggesting a functional role of this receptor. As a sign of TLR4 activation, an LPS challenge increased IL-6 release in normal and tumoral cells. By flow cytometry, TLR4 baseline expression was revealed at the plasma membrane of tumoral lactotrophs, without changes noted in the percentage of double PRL/TLR4 positive cells after LPS stimulus. Increases in TLR4 intracellular expression were detected as well as rises in CD14, p-Akt and NF-κB after an LPS challenge, as assessed by western blotting. The TLR4/PRL and PRL/NF-κB co-localization was also corroborated by immunofluorescence and the involvement of PI3K/Akt signaling in lactotroph proliferation and IL-6 release was revealed through the PI3K inhibitor Ly-294002. In addition, 17ß-estradiol attenuated the LPS-evoked increase in tumoral lactotroph proliferation and IL-6 release. Collectively these results demonstrate the presence of functional TLR4 in lactotrophs from estrogen-induced hyperplasic pituitaries, which responded to the proliferative stimulation and IL-6 release induced by LPS through TLR4/CD14, with a contribution of the PI3K-Akt and NF-κB signaling pathways.


Subject(s)
Cell Proliferation/drug effects , Lipopolysaccharides/pharmacology , Pituitary Gland/metabolism , Pituitary Neoplasms/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Hyperplasia/metabolism , Interleukin-6/metabolism , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Gland/ultrastructure , Pituitary Neoplasms/immunology , Pituitary Neoplasms/ultrastructure , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology
13.
Toxicol Appl Pharmacol ; 240(1): 55-65, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19595700

ABSTRACT

Bromocriptine (Bc) produces pituitary tumoral mass regression which induces the cellular death that was classically described as apoptosis. However, recent works have related that other mechanisms of cell death could also be involved in the maintenance of physiological and pathological pituitary homeostasis. The aim of this study was to evaluate and characterize the different types of cell death in the involution induced by Bc in experimental rat pituitary tumors. The current study demonstrated that Bc induced an effective regression of estrogen induced pituitary tumors by a mechanism identified as parapoptosis. This alternative cell death was ultrastructurally recognized by extensive cytoplasmic vacuolization and an increased cell electron density, represented around 25% of the total pituitary cells counted. Furthermore, the results obtained from biochemical assays did not correspond to the criteria of apoptosis or necrosis. We also investigated the participation of p38, ERK1/2 and PKC delta in the parapoptotic pathway. An important observation was the significant increase in phosphorylated forms of these MAPKs, the holoenzyme and catalytic fragments of PKC delta in nuclear fractions after Bc administration compared to control and estrogen treated rats. Furthermore, the immunolocalization at ultrastructural level of these kinases showed a similar distribution pattern, with a prevalent localization at nuclear level in lactotrophs from Bc treated rats. In summary, we determined that parapoptosis is the predominant cell death type involved in the regression of pituitary tumors in response to Bc treatment, and may cause the activation of PKC delta, ERK1/2 and p38.


Subject(s)
Apoptosis/drug effects , Bromocriptine/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/pathology , Prolactinoma/drug therapy , Prolactinoma/pathology , Animals , Apoptosis/physiology , Cell Death/drug effects , Cell Death/physiology , MAP Kinase Kinase 2/physiology , Male , Mitogen-Activated Protein Kinase 3/physiology , Pituitary Neoplasms/enzymology , Prolactinoma/enzymology , Protein Kinase C-delta/physiology , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases/physiology
14.
J Mol Histol ; 40(5-6): 417-25, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20177957

ABSTRACT

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


Subject(s)
Lactotrophs/enzymology , Lactotrophs/pathology , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Lactotrophs/drug effects , Lactotrophs/ultrastructure , Mitogens/pharmacology , Nuclear Envelope/drug effects , Nuclear Envelope/enzymology , Nuclear Envelope/ultrastructure , Pituitary Neoplasms/enzymology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/ultrastructure , Protein Kinase C-alpha/ultrastructure , Protein Kinase C-epsilon/ultrastructure , Protein Transport/drug effects , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology
15.
Mol Cell Endocrinol ; 289(1-2): 77-84, 2008 Jul 16.
Article in English | MEDLINE | ID: mdl-18534741

ABSTRACT

The aim of this investigation was to contribute to current knowledge about intracellular mechanisms that are involved in lactotroph cell proliferation, by evaluating the role of PKCalpha, PKCepsilon and extracellular-signal regulated kinase (ERK) 1/2 in response to phorbol 12-myristate13-acetate (PMA). In primary pituitary cultures, the activation of protein kinase C (PKC) by PMA for 15 min stimulated lactotroph proliferation; whereas a prolonged activation for 3-8h diminished this proliferative effect. The use of PMA for 15 min-activated PKCepsilon and ERK1/2, whereas incubation with PMA for 3 h induced PKCalpha activation and attenuated the PMA-triggered phosphorylation of ERK1/2. The following inhibitors: PKCs (bisindolylmaleimide I), PKCepsilon (epsilonV1 peptide) and ERK1/2 (PD98059) prevented the mitogenic activity induced by PMA for 15 min. Lactotroph cells stimulated with PMA for 15 min showed a translocation of PKCepsilon to membrane compartment and nucleus. These results thus establish that PKCepsilon plays an essential role in the lactotroph proliferation induced by PMA by triggering signals that involve ERK1/2 activation.


Subject(s)
Cell Proliferation/drug effects , Lactotrophs/cytology , Lactotrophs/physiology , Protein Kinase C-epsilon/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Female , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport/drug effects , Rats , Rats, Wistar
16.
Histochem Cell Biol ; 129(5): 631-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18299881

ABSTRACT

The objectives of the present work were to assess whether epithelial cells from the different segments of epididymis express TR alpha 1-beta 1 isoforms, to depict its subcellular immunolocalization and to evaluate changes in their expression in rats experimentally submitted to a hypothyroid state by injection of 131I. In euthyroid and hypothyroid groups, TR protein was expressed in epididymal epithelial cells, mainly in the cytoplasmic compartment while only a few one showed a staining in the nucleus as well. A similar TR immunostaining pattern was detected in the different segments of the epididymis. In hypothyroid rats, the number of TR-immunoreactive epithelial cells as well as the intensity of the cytoplasmic staining significantly increased in all sections analyzed. In consonance to the immunocytochemical analysis, the expression of TR alpha 1-beta 1 isoforms, assessed by Western blot revealed significantly higher levels of TR in cytosol compared to the nuclear fractions. Furthermore, TR expression of both alpha 1 and beta 1 isoforms and their mRNA levels were increased by the hypothyroid state. The immuno-electron-microscopy showed specific reaction for TR in principal cells associated with eucromatin, cytosolic matrix and mitochondria. The differences in expression levels assessed in control and thyroidectomized rats ascertain a specific function of TH on this organ.


Subject(s)
Epididymis/metabolism , Epithelial Cells/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Animals , Blotting, Western , Cell Nucleus/metabolism , Cytoplasm/metabolism , Epididymis/pathology , Epididymis/ultrastructure , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Gene Expression , Hypothyroidism/genetics , Hypothyroidism/metabolism , Hypothyroidism/physiopathology , Immunohistochemistry , Male , Microscopy, Immunoelectron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Thyroid Hormone/analysis , Receptors, Thyroid Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/metabolism , Thyroid Gland/physiopathology , Thyroid Hormone Receptors alpha/analysis , Thyroid Hormone Receptors beta/analysis , Thyroxine/blood , Triiodothyronine/blood
17.
Fertil Steril ; 89(6): 1613-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17531233

ABSTRACT

OBJECTIVE: Evaluation of uteroglobin (UG) expression in the fallopian tube in different tubal diseases. DESIGN: The UG was screened and quantified in samples of fallopian tubes from patients with salpingitis, hydrosalpinx, and ectopic pregnancy by exposing the UG with immunohistochemical techniques. SETTING: University hospital and electron microscopy center. PATIENT(S): Women with pelvic inflammatory disease (PID) and complicated tubal ectopic pregnancy consulting for medical care. INTERVENTION(S): Salpingectomy. MAIN OUTCOME MEASURE(S): Tubal tissues were collected and examined using regular pathologic techniques. The UG immunoreactivity in the tubal epithelium was also assessed. RESULT(S): Fallopian tube epithelium displayed an increased UG expression in patients with PID and complicated tubal pregnancy compared with control patients. CONCLUSION(S): Uteroglobin is present in the human fallopian tube as a secretory protein and appears to be involved in immunosuppressive responses in the fallopian tube.


Subject(s)
Fallopian Tubes/metabolism , Inflammation/etiology , Pregnancy, Ectopic/genetics , Uteroglobin/metabolism , Adult , Fallopian Tubes/immunology , Fallopian Tubes/surgery , Female , Humans , Immunohistochemistry , Pregnancy , Salpingitis/surgery
18.
Cell Tissue Res ; 327(1): 121-32, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17024419

ABSTRACT

We have investigated the expression of receptors for insulin and insulin-like growth factor 1 (IGF-1) in rat pituitary cells in vitro and examined the morphological and proliferative changes induced in adenohypophyseal cells by insulin and IGF-1. The proliferation of lactotrophs was determined by double-immunostaining for bromodeoxyuridine and prolactin. Incubation with insulin (10, 100 or 1000 ng/ml) or IGF-1 (5, 30 or 100 ng/ml) for 48 or 72 h significantly increased the number of lactotrophs undergoing mitosis. Co-incubation of insulin or IGF-1 with genistein (25 microM), an inhibitor of the tyrosine kinase receptor, reduced the proliferation of lactotrophs elicited by the hormone and the growth factor. The receptors for insulin and IGF-1 were localized in intact pituitary cells by ultrastructural immunocytochemistry with the colloidal gold-protein A technique. Gonadotrophs expressed both receptors, specific labelling being restricted to this cell type. Electron-microscopical observations of pituitary cell cultures incubated with insulin or IGF-1 revealed gonadotroph cells exhibiting the fine-structural features of enhanced protein synthetic activity. These findings suggest that both insulin and IGF-1 are able to induce the proliferation of lactotrophs through an indirect mechanism mediated by a factor synthesized by gonadotroph cells, in addition to stimulating the biosynthetic activity of the gonadotroph in a direct manner.


Subject(s)
Gonadotrophs/cytology , Lactotrophs/cytology , Microscopy, Electron, Transmission/methods , Pituitary Gland, Anterior/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Animals , Cell Count , Cell Proliferation , Cells, Cultured , DNA/biosynthesis , Dose-Response Relationship, Drug , Drug Combinations , Female , Fluorescent Antibody Technique, Direct , Gonadotrophs/metabolism , Gonadotrophs/ultrastructure , Immunoenzyme Techniques , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Lactotrophs/metabolism , Lactotrophs/ultrastructure , Pituitary Gland, Anterior/ultrastructure , Rats , Rats, Wistar , Receptor, IGF Type 1/ultrastructure , Receptor, Insulin/ultrastructure
19.
Histochem Cell Biol ; 124(3-4): 291-301, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16133120

ABSTRACT

The effects of IGF-1, 17 beta oestradiol and its functional interaction on lactotrophs cell proliferation were evaluated. In addition we investigated the involvement of PKC alpha, epsilon and phosphorilated ERK, in the mitogenic process. Primary cell cultures of adenohypophysis from female Wistar rats were studied in serum free conditions. The proliferation of lactotrophs was determined by double immunostaining for BrdU and PRL. The incubation with IGF-1 5, 30 or 100 ng/ml during 48 or 72 h increased lactotrophs proliferation two-threefold depending on IGF-1 concentration. Co-incubation of IGF-1 (30 ng/ml) with genistein (25 microM) or BIM (0.5 or 2 microM), lowered of tyrosine kinase receptor or of PKC respectively, inhibited the induced IGF-1 lactotrophs proliferation. 17 beta oestradiol (1, 10 or 100 nM) had not mitogenic effect, whereas in the presence of serum PRL cells proliferation was stimulated. Co-incubation with 1 nM oestradiol and IGF-1 significantly decreased the lactotroph BrdU-labelling achieved with IGF-1. PKC alpha, epsilon and ERK1/2 levels measured by western blot augmented in the presence of IGF-1 and were inhibited with the addition of genistein, supporting a participation of these enzymes in the proliferate process. Co-incubation of IGF-1 with 1 nM oestradiol decreased both PKC isoforms and activated ERK1/2 levels, suggesting that oestradiol would exert its antiproliferative effect by acting on the signalling pathway of IGF-1. The results revealed antagonic effects of oestradiol on lactotroph proliferation depending on its concentration and the presence of IGF-1.


Subject(s)
Estradiol/physiology , Insulin-Like Growth Factor I/physiology , Pituitary Gland, Anterior/cytology , Animals , Cell Proliferation , Cells, Cultured , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Phosphorylation , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/ultrastructure , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/metabolism , Rats , Rats, Wistar
20.
J Mol Histol ; 36(8-9): 447-54, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16733790

ABSTRACT

Vascular endothelial growth factor (VEGF) is an important angiogenic factor in the pituitary gland. The objective of this study was to unveil the VEGF subcellular localisation in different pituitary cell types and to evaluate changes in its expression at different time intervals after oestrogen stimulation. A relevant feature demonstrated was the identification of this cytokine in the nucleus and cytoplasm of lactotrophs, somatotrophs and gonadotrophs, as well as in follicle-stellate cells of male rats. Oestrogen treatment increased the number of VEGF immunopositive cells and its expression detected differentially by western blot in both nucleus and cytoplasm of pituitary cells when compared to the control. At ultrastructural level VEGF appeared associated with nucleolus and euchromatin involving a possible internal autocrine loop. In lactotrophs, the predominant cell of the tumour, VEGF was immunodetected in RER, Golgi complex, and vesicular organelles, supporting further the association with an auto-paracrine effect exerted by VEGF. The nucleus/cytoplasm ratio of VEGF revealed a prevalent accumulation of VEGF in the cytoplasm. The presence of VEGF in the nucleus may probably be associated with a translocation to this cell compartment. This study demonstrated a cytoplasmic and nuclear immunolocalisation of VEGF in normal and tumoural adenohypophyseal cells. In the course of prolactinoma development, the oestrogen stimulated VEGF expression in tumoural cells, promoting a vascular adaptation which contributes to growth and progression of the tumour.


Subject(s)
Estrogens/pharmacology , Lactotrophs/metabolism , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Cell Extracts , Cell Nucleus/drug effects , Cytoplasm/metabolism , Immunohistochemistry , Lactotrophs/cytology , Lactotrophs/ultrastructure , Male , Pituitary Neoplasms/chemically induced , Prolactinoma/chemically induced , Protein Transport/drug effects , Rats , Rats, Wistar , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...