Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Intervalo de año de publicación
1.
Food Chem Toxicol ; 158: 112649, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34728246

RESUMEN

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.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Disruptores Endocrinos/toxicidad , Hipófisis , Efectos Tardíos de la Exposición Prenatal , Receptores de Estrógenos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Lactotrofos/efectos de los fármacos , Lactotrofos/metabolismo , Masculino , Hipófisis/citología , Hipófisis/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas , Ratas Wistar , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo
2.
J Cell Physiol ; 236(10): 6974-6987, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33682941

RESUMEN

Octreotide (OCT) is used to inhibit hormone secretion and growth in somatotroph tumors, although a significant percentage of patients are resistant. It has also been tested in nonfunctioning (NF) tumors but with poor results, with these outcomes having been associated with SSTR2 levels and impaired signaling. We investigated whether OCT inhibitory effects can be improved by TGF-ß1 in functioning and nonfunctioning somatotroph tumor cells. OCT effects on hormone secretion and proliferation were analyzed in the presence of TGF-ß1 in WT and SSTR2-overexpressing secreting GH3 and silent somatotroph tumor cells. The mechanism underlying these effects was assessed by studying SSTR and TGFßR signaling pathways mediators. In addition, we analyzed the effects of OCT/TGF-ß1 treatment on tumor growth and cell proliferation in vivo. The inhibitory effects of OCT on GH- and PRL-secretion and proliferation were improved in the presence of TGF-ß1, as well as by SSTR2 overexpression. The OCT/TGF-ß1 treatment induced downregulation of pERK1/2 and pAkt, upregulation of pSmad3, and inhibition of cyclin D1. In vivo experiments showed that OCT in the presence of TGF-ß1 blocked tumor volume growth, decreased cell proliferation, and increased tumor necrosis. These results indicate that SSTR2 levels and the stimulation of TGF-ß1/TGFßR/Smad2/3 pathway are important for strengthening the antiproliferative and antisecretory effects of OCT.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Proliferación Celular/efectos de los fármacos , Octreótido/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Somatotrofos/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología , Animales , Línea Celular , Femenino , Humanos , Ratones Desnudos , Fosforilación , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Ratas , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal , Somatotrofos/metabolismo , Somatotrofos/patología , Carga Tumoral/efectos de los fármacos
3.
Chemosphere ; 258: 127304, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32559490

RESUMEN

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.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Ácidos Ftálicos/toxicidad , Receptores de Estrógenos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Dietilhexil Ftalato/metabolismo , Disruptores Endocrinos/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Lactotrofos/efectos de los fármacos , Lactotrofos/metabolismo , Hipófisis/efectos de los fármacos , Ratas
4.
Mol Neurobiol ; 56(3): 1539-1557, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29948953

RESUMEN

Chronic metabolic alterations may represent a risk factor for the development of cognitive impairment, dementia, or neurodegenerative diseases. Hyperglycemia and obesity are known to imprint epigenetic markers that compromise the proper expression of cell survival genes. Here, we showed that chronic hyperglycemia (60 days) induced by a single intraperitoneal injection of streptozotocin compromised cognition by reducing hippocampal ERK signaling and by inducing neurotoxicity in rats. The mechanisms appear to be linked to reduced active DNA demethylation and diminished expression of the neuroprotective transcription factor REST. The impact of the relationship between adiposity and DNA hypermethylation on REST expression was also demonstrated in peripheral blood mononuclear cells in obese children with reduced levels of blood ascorbate. The reversible nature of epigenetic modifications and the cognitive impairment reported in obese children, adolescents, and adults suggest that the correction of the anthropometry and the peripheral metabolic alterations would protect brain homeostasis and reduce the risk of developing neurodegenerative diseases.


Asunto(s)
Trastornos del Conocimiento/etiología , Diabetes Mellitus Experimental/complicaciones , Hipocampo/metabolismo , Hiperglucemia/complicaciones , Proteínas Represoras/metabolismo , Animales , Reacción de Prevención/fisiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Metilación de ADN , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Aprendizaje por Laberinto/fisiología , Ratas , Proteínas Represoras/genética
5.
J Endocrinol ; 240(2): 229-241, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30400032

RESUMEN

The molecular mechanisms underlying the ERα nuclear/cytoplasmic pool that modulates pituitary cell proliferation have been widely described, but it is still not clear how ERα is targeted to the plasma membrane. The aim of this study was to analyse ERα palmitoylation and the plasma membrane ERα (mERα) pool, and their participation in E2-triggered membrane-initiated signalling in normal and pituitary tumour cell growth. Cell cultures were prepared from anterior pituitaries of female Wistar rats and tumour GH3 cells, and treated with 10 nM of oestradiol (E2). The basal expression of ERα was higher in tumour GH3 than in normal pituitary cells. Full-length palmitoylated ERα was observed in normal and pituitary tumour cells, demonstrating that E2 stimulation increased both, ERα in plasma membrane and ERα and caveolin-1 interaction after short-term treatment. In addition, the Dhhc7 and Dhhc21 palmitoylases were negatively regulated after sustained stimulation of E2 for 3 h. Although the uptake of BrdU into the nucleus in normal pituitary cells was not modified by E2, a significant increase in the GH3 tumoural cell, as well as ERK1/2 activation, with this effect being mimicked by PPT, a selective antagonist of ERα. These proliferative effects were blocked by ICI 182780 and the global inhibitor of palmitoylation. These findings indicate that ERα palmitoylation modulated the mERα pool and consequently the ERK1/2 pathway, thereby contributing to pituitary tumour cell proliferation. These results suggest that the plasma membrane ERα pool might be related to the proliferative behaviour of prolactinoma and may be a marker of pituitary tumour growth.


Asunto(s)
Membrana Celular/metabolismo , Proliferación Celular , Receptor alfa de Estrógeno/metabolismo , Neoplasias Hipofisarias/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Células Cultivadas , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Femenino , Fulvestrant/farmacología , Expresión Génica/efectos de los fármacos , Lipoilación/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Ratas Wistar
6.
Free Radic Biol Med ; 120: 41-55, 2018 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-29548793

RESUMEN

The cellular transformation of normal functional cells to neoplastic ones implies alterations in the cellular metabolism and mitochondrial function in order to provide the bioenergetics and growth requirements for tumour growth progression. Currently, the mitochondrial physiology and dynamic shift during pituitary tumour development are not well understood. Pituitary tumours present endocrine neoplastic benign growth which, in previous reports, we had shown that in addition to increased proliferation, these tumours were also characterized by cellular senescence signs with no indication of apoptosis. Here, we show clear evidence of oxidative stress in pituitary cells, accompanied by bigger and round mitochondria during tumour development, associated with augmented biogenesis and an increased fusion process. An activation of the Nrf2 stress response pathway together with the attenuation of the oxidative damage signs occurring during tumour development were also observed which will probably provide survival advantages to the pituitary cells. These neoplasms also presented a progressive increase in lactate production, suggesting a metabolic shift towards glycolysis metabolism. These findings might imply an oxidative stress state that could impact on the pathogenesis of pituitary tumours. These data may also reflect that pituitary cells can modulate their metabolism to adapt to different energy requirements and signalling events in a pathophysiological situation to obtain protection from damage and enhance their survival chances. Thus, we suggest that mitochondria function, oxidative stress or damage might play a critical role in pituitary tumour progression.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Neoplasias Hipofisarias/metabolismo , Adaptación Fisiológica/fisiología , Animales , Antioxidantes/metabolismo , Metabolismo Energético/fisiología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
7.
J Cell Physiol ; 233(2): 1402-1413, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28542730

RESUMEN

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.


Asunto(s)
Proliferación Celular , Receptor beta de Estrógeno/metabolismo , Ciclo Estral/metabolismo , Lactotrofos/enzimología , Fosfohidrolasa PTEN/metabolismo , Somatotrofos/enzimología , Animales , Células Cultivadas , Estradiol/metabolismo , Estradiol/farmacología , Receptor beta de Estrógeno/agonistas , Receptor beta de Estrógeno/genética , Terapia de Reemplazo de Estrógeno , Femenino , Fase G1 , Lactotrofos/efectos de los fármacos , Masculino , Nitrilos/farmacología , Ovariectomía , Ratas Wistar , Transducción de Señal , Somatotrofos/efectos de los fármacos , Transfección
8.
J Photochem Photobiol B ; 153: 445-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26569453

RESUMEN

Several research efforts have been focused on finding newer and more efficient photosensitizers for photodynamic therapy (PDT). Although, it was demonstrated that riboflavin is an efficient photosensitizer for PDT, the effect of its ester derivate, riboflavin 2',3',4',5'-tetraacetate (RFTA), which has higher cellular uptake, has not been well defined. To evaluate the cell death generated by applying RFTA as the photosensitizer in PDT in a human cancer cell line of squamous carcinoma (SCC-13), these cells were incubated with riboflavin and its ester derivate, RFTA followed by irradiation with different blue light doses. Cell viability was evaluated using neutral red uptake assay and cell death was evaluated using transmission electron microscopy, TUNEL assay and annexin V-PE/7AAD double staining. The expression of caspase-3, Bax, Bcl-2, ERK 1/2 and p38(MAPK) was evaluated by Western blotting and generation of intracellular ROS and changes in anion superoxide levels were analyzed using 2',7'-dichlorofluorescein-diacetate and dihydroethidium dye, respectively. RFTA-PDT generated a decrease in cancer cell viability in a light dose-response. Treated SCC-13 cells exhibited chromatin condensation, formation of apoptotic bodies, increases in TUNEL-positive cells, phosphatidylserine externalization and decreased procaspase-3 and Bcl-2 protein expression and increment of ERK 1/2 phosphorylation. Moreover, trolox abolished the effect of PDT on cell viability linking the increase in intracellular ROS levels with the cell death observed, whereas that the pre-treatment with MEK inhibitor did not induce changes in SCC-13 cell survival. These findings demonstrate the effects of RFTA in triggering apoptosis induced by ROS (\O2(-)) production after visible light irradiation of squamous carcinoma cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Riboflavina/análogos & derivados , Riboflavina/farmacología , Apoptosis/efectos de la radiación , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Caspasa 3/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Humanos , Luz , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Riboflavina/química , Riboflavina/uso terapéutico , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Mol Cell Endocrinol ; 415: 100-13, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26282612

RESUMEN

Considering that the role of ERß in the growth of pituitary cells is not well known, the aim of this work was to determine the expression of ERß in normal and tumoral cells and to investigate its implications in the proliferative control of this endocrine gland, by analyzing the participation of cyclin D1, Cdk4 and p21. Our results showed that the expression of ERß decreased during pituitary tumoral development induced by chronic E2 stimulation. The 20 ± 1.6% of normal adenohypophyseal cells expressed ERß, with this protein being reduced in the hyperplastic/adenomatous pituitary: at 20 days the ERß+ population was 10.7 ± 2.2%, while after 40 and 60 days of treatment an almost complete loss in the ERß expression was observed (40 d: 1 ± 0.6%; 60 d: 2 ± 0.6%). The ERα/ß ratio increased starting from tumors at 40 days, mainly due to the loss of ERß expression. The cell proliferation was analyzed in normal and hyperplastic pituitary and also in GH3ß- and GH3ß+ which contained different levels of ERß expression, and therefore different ERα/ß ratios. The over-expression of ERß inhibited the GH3 cell proliferation and expression of cyclin D1 and ERα. Also, the ERß activation by its agonist DPN changed the subcellular localization of p21, inducing an increase in the p21 nuclear expression, where it acts as a tumoral suppressor. These results show that ERß exerts an inhibitory role on pituitary cell proliferation, and that this effect may be partially due to the modulation of some key regulators of the cell cycle, such as cyclin D1 and p21. These data contribute significantly to the understanding of the ER effects in the proliferative control of pituitary gland, specifically related to the ERß function in the E2 actions on this endocrine gland.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Estradiol/efectos adversos , Receptor beta de Estrógeno/metabolismo , Adenohipófisis/citología , Neoplasias Hipofisarias/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , NAD/farmacología , Adenohipófisis/metabolismo , Neoplasias Hipofisarias/inducido químicamente , Neoplasias Hipofisarias/patología , Ratas
11.
Int J Pharm ; 478(1): 258-267, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25448587

RESUMEN

Large oral doses of ACZ lower the intraocular pressure (IOP), but usually lead to a multitude of systemic side effects, including gastrointestinal upset. The present study was undertaken to evaluate the effect of ACZ on the histological structure of rat duodenal mucosa and to assess a possible protective role of the complex formation of ACZ with HP-ß-CD, either separately or in combination with a third compound, on the gut epithelial layer by histological and ultrastructural examinations of sections of rat duodenum exposed to ACZ or its formulations. In addition, the transport process of ACZ and its binary or ternary complexes across the duodenal mucosa by means of the single-pass intestinal perfusion (SPIP) method in rats was evaluated. Evidence was found that ACZ alters intestinal permeability and induces damage to the rat small intestine. In contrast, ACZ-induced intestinal injury may be abrogated by ACZ complexation. In addition, the complexation of ACZ with HP-ß-CD, alone or in combination with a third compound, facilitated significant levels of ACZ uptake across the rat duodenal segment. Ternary complexes of ACZ with HP-ß-CD in combination with TEA (triethanolamine) or calcium ions were found to provide an excellent approach that enabled an increased apparent permeability of ACZ across the duodenal epithelium, with a concomitant ability to preserve the integrity of the gut epithelium from ACZ-induced injury. These results could be useful for the design and development of novel ACZ formulations that can reduce GI toxicity, while still maintaining their essential therapeutic efficacies.


Asunto(s)
Acetazolamida , beta-Ciclodextrinas , 2-Hidroxipropil-beta-Ciclodextrina , Acetazolamida/administración & dosificación , Acetazolamida/química , Acetazolamida/farmacocinética , Acetazolamida/toxicidad , Animales , Calcio/administración & dosificación , Calcio/química , Calcio/farmacocinética , Calcio/toxicidad , Duodeno/efectos de los fármacos , Duodeno/patología , Duodeno/ultraestructura , Etanolaminas/administración & dosificación , Etanolaminas/química , Etanolaminas/farmacocinética , Etanolaminas/toxicidad , Absorción Intestinal , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/ultraestructura , Masculino , Microscopía Electrónica de Transmisión , Ratas Wistar , beta-Ciclodextrinas/administración & dosificación , beta-Ciclodextrinas/química , beta-Ciclodextrinas/farmacocinética , beta-Ciclodextrinas/toxicidad
12.
Mol Neurobiol ; 51(3): 1353-67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25037702

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is an animal model that mimics many of the clinical and pathological features of the human disease multiple sclerosis (MS). Both are inflammatory demyelinating and neurodegenerative pathologies of the central nervous system associated with motor, sensory, and cognitive deficits. In MS, gray matter atrophy is related to the emergence of cognitive deficits and contributes to clinical progression. In particular, prefrontal cortex injury and dysfunction have been correlated to the development of fatigue, one of the most common and disabling symptoms in MS. However, the molecular bases of these changes remain unknown. Taking advantage of EAE similitude, we herein analyze functional and morphological changes in isolated cortical presynaptic terminals (synaptosomes) from an acute rat model. We found impaired glutamate release in the frontal cortex from EAE rats. This defect appeared along with the onset of the disease, reversing when clinical signs were no more evident. Biochemical analysis of EAE synaptosomes revealed alterations in the presynaptic release machinery and in the response to depolarization, which was accompanied by abnormal synapsin I phosphorylation and dispersion. These changes were associated with reduced synaptic vesicle mobility, with no alterations in synaptosomal morphology as evidenced by electron microscopy. The present are the first pieces of evidence unraveling the molecular mechanisms of frontal cortex neuronal dysfunction in EAE and, possibly, MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Lóbulo Frontal/efectos de los fármacos , Ácido Glutámico/farmacología , Sinaptosomas/efectos de los fármacos , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Lóbulo Frontal/metabolismo , Ácido Glutámico/administración & dosificación , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Ratas Wistar , Sinapsinas/metabolismo , Sinaptosomas/metabolismo
13.
Int J Exp Pathol ; 94(6): 399-411, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23998365

RESUMEN

Bronchiolar Clara cells play a critical role in lung homoeostasis. The main goal of this study was to evaluate the effects of chronic allergy on these cells and the efficacy of budesonide (BUD) and montelukast (MK) in restoring their typical phenotypes after ovalbumin-induced chronic allergy in mice. Chronic allergy induced extensive bronchiolar Alcian blue-periodic acid-Schiff (AB/PAS)-positive metaplasia. In addition, cells accumulated numerous big electron-lucent granules negative for Clara cell main secretory protein (CC16), and consequently, CC16 was significantly reduced in bronchoalveolar lavage. A concomitant reduction in SP-D and CYP2E1 content was observed. The phenotypic changes induced by allergy were pharmacologically reversed by both treatments; MK was more efficient than BUD in doing so. MK decreased AB/PAS reactivity to control levels whereas they remained persistently elevated after BUD. Moreover, most non-ciliated cells recovered their normal morphology after MK, whereas for BUD normal cells coexisted with 'transitional' cells that contained remnant mucous granules and stained strongly for CC16 and SP-D. Glucocorticoids were also less able to reduce inflammatory infiltration and maintained higher percentage of neutrophils, which may have contributed to prolonged mucin expression. These results show that chronic allergy-induced mucous metaplasia of Clara cells affects their defensive mechanisms. However, anti-inflammatory treatments were able to re-establish the normal phenotype of Clara cell, with MK being more efficient at restoring a normal profile than BUD. This study highlights the role of epithelial cells in lung injuries and their contribution to anti-inflammatory therapies.


Asunto(s)
Acetatos/uso terapéutico , Asma/tratamiento farmacológico , Asma/patología , Bronquios/patología , Budesonida/uso terapéutico , Fenotipo , Quinolinas/uso terapéutico , Acetatos/farmacología , Animales , Antiasmáticos/farmacología , Antiasmáticos/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Asma/inducido químicamente , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Budesonida/farmacología , Enfermedad Crónica , Ciclopropanos , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Femenino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/efectos adversos , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Quinolinas/farmacología , Sulfuros , Uteroglobina/metabolismo
14.
Am J Physiol Endocrinol Metab ; 305(1): E41-9, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23651845

RESUMEN

In the present work, we investigated the effect of 17ß-estradiol (E2) and basic fibroblast growth factor 2 (FGF2) on the lactotroph cell-proliferative response and the related membrane-initiated signaling pathway. Anterior pituitary mixed-cell cultures of random, cycling 3-mo-old female rats were treated with 10 nM E2, E2 membrane-impermeable conjugated BSA (E2-BSA), PPT (ERα agonist), and DPN (ERß agonist) alone or combined with FGF2 (10 ng/ml) for 30 min or 4 h. Although our results showed that the uptake of BrdU into the nucleus of lactotrophs was not modified by E2 or FGF2 alone, a significant increase in the lactotroph uptake of BrdU was observed after E2/FGF2 coincubation, with this effect being mimicked by PPT/FGF2. These proliferative effects were blocked by ICI 182,780 or PD-98059. The involvement of membrane ER in the proliferative response of prolactin cells induced by the steroid and FGF2 coincubation was confirmed using E2-BSA, and the association between ERα and FGF receptor was observed after E2/FGF2 treatment by immunoprecipitation. A significant increase in the ERK1/2 expression was noted after E2, E2-BSA, PPT, and FGF2 alone, which was more noticeable after E2-BSA/FGF2, E2/FGF2, or PPT/FGF2 treatments. This study provides evidence that E2 and FGF2 exert a cooperative effect on the lactotroph proliferation principally by signaling initiated at the plasma membrane triggering a genomic effect mediated by MEK/ERK1/2, a common signaling pathway, that finally regulates the lactotroph population, thus contributing to pituitary plasticity.


Asunto(s)
Membrana Celular/metabolismo , Estradiol/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Lactotrofos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Adenohipófisis/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Lactotrofos/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Adenohipófisis/citología , Cultivo Primario de Células , Ratas , Ratas Wistar
19.
Mol Cell Endocrinol ; 355(1): 169-79, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22366173

RESUMEN

In the present work we aimed at identifying ERα in the plasma membrane of normal anterior pituitary cells and investigated if 17ß-estradiol was able to induce their subcellular redistribution. Our results show that about 8% of anterior pituitary cells expressed ERα in the plasma membrane, with the geometrical mean fluorescence intensity being increased after steroid hormone treatment. 17ß-Estradiol and the selective ERα agonist PPT induced an increase of ERα expression in the plasma membrane and activated the PKCα/ERK 1/2 pathway in a time-course not compatible with genomic actions, thus supporting the notion of membrane-initiated effects. These findings suggest that 17ß-estradiol stimulates the translocation of endogenous ERα to the plasma membrane, consequently modulating this ER pool and leading to cellular biological effects in normal anterior pituitary gland.


Asunto(s)
Membrana Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Adenohipófisis/efectos de los fármacos , Animales , Membrana Celular/metabolismo , Células Cultivadas , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Ligandos , Fenoles , Adenohipófisis/citología , Adenohipófisis/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Transporte de Proteínas/efectos de los fármacos , Pirazoles/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
20.
Am J Physiol Endocrinol Metab ; 302(10): E1189-97, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22354782

RESUMEN

Considering that estradiol is a major modulator of prolactin (PRL) secretion, the aim of the present study was to analyze the role of membrane estradiol receptor-α (mERα) in the regulatory effect of this hormone on the PRL secretion induced by thyrotropin-releasing hormone (TRH) by focusing on the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway activation. Anterior pituitary cell cultures from female rats were treated with 17ß-estradiol (E(2), 10 nM) and its membrane-impermeable conjugated estradiol (E(2)-BSA, 10 nM) alone or coincubated with TRH (10 nM) for 30 min, with PRL levels being determined by RIA. Although E(2), E(2)-BSA, TRH, and E(2)/TRH differentially increased the PRL secretion, the highest levels were achieved with E(2)-BSA/TRH. ICI-182,780 did not modify the TRH-induced PRL release but significantly inhibited the PRL secretion promoted by E(2) or E(2)-BSA alone or in coincubation with TRH. The PI3K inhibitors LY-294002 and wortmannin partially inhibited the PRL release induced by E(2)-BSA, TRH, and E(2)/TRH and totally inhibited the PRL levels stimulated by E(2)-BSA/TRH, suggesting that the mER mediated the cooperative effect of E(2) on TRH-induced PRL release through the PI3K pathway. Also, the involvement of this kinase was supported by the translocation of its regulatory subunit p85α from the cytoplasm to the plasma membrane in the lactotroph cells treated with E(2)-BSA and TRH alone or in coincubation. A significant increase of phosphorylated Akt was induced by E(2)-BSA/TRH. Finally, the changes of ERα expression in the plasmalemma of pituitary cells were examined by confocal microscopy and flow cytometry, which revealed that the mobilization of intracellular ERα to the plasma membrane of lactotroph cells was only induced by E(2). These finding showed that E(2) may act as a modulator of the secretory response of lactotrophs induced by TRH through mER, with the contribution by PI3K/Akt pathway activation providing a new insight into the mechanisms underlying the nongenomic action of E(2) in the pituitary.


Asunto(s)
Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Adenohipófisis , Prolactina/metabolismo , Transducción de Señal/fisiología , Hormona Liberadora de Tirotropina/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas de la Membrana/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Albúmina Sérica Bovina/farmacología , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...