Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
J Prev Med Hyg ; 60(3): E229-E236, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31650059

ABSTRACT

INTRODUCTION: Hospitals are known to be the most complex entities to manage. In fact, the main problem in healthcare are the expensive needs with limited resources. During the last years the complexity of the nurse manager role has gradually changed from assistance to management. However, nowadays the methods for quantifying the nurse managers' skills and performance are not available. The aim of this study is to implement a method to assess and measure the skills of the nurse managers. An innovative indicator to globally evaluate the features, the professional skills and their performance is described. METHODS: The authors started with an interview with the directors of all the nurses as the top experts of the nurse managers' technical skills. The purpose of this step was to understand what were the features of a valuable nurse manager. The methods identified three different aspects (qualitative, quantitative and relational) that were transformed in a single indicator. These parameters also enable to identify the strengths and weaknesses of each professional. An important implication of this score is the possible improvement of loss-making skills. RESULTS: A total of 18 centres, with their nurse managers, were evaluated in this study. All the results confirmed the judgment of the Healthcare Professions Structure Manager. CONCLUSIONS: This assessment method, validated with these tests, evaluated the nurse manager's ability to deal with personnel, resources and patients and to quantify his/her organizational and welfare performances. It is useful for planning actions that allow nurse managers to improve their skills.


Subject(s)
Hospital Administration/standards , Nurse Administrators/standards , Professional Competence , Work Performance , Hospitals , Humans , Quality Indicators, Health Care
2.
Diabetes Obes Metab ; 18(5): 537-41, 2016 May.
Article in English | MEDLINE | ID: mdl-26847506

ABSTRACT

Gliptins are anti-type 2 diabetes (T2D) drugs that regulate glycaemia by preventing endogenous glucagon-like peptide-1 (GLP-1) degradation. Chronically administered gliptins before experimental stroke can also induce neuroprotection, and this effect is potentially relevant for reducing brain damage in patients with T2D and high risk of stroke. It is not known, however, whether acute gliptin treatment after stroke (mimicking a post-hospitalization treatment) is neuroprotective or whether gliptin-mediated neuroprotection occurs via GLP-1-receptor (GLP-1R) activation. To answer these two questions, wild-type and glp-1r(-/-) mice were subjected to transient middle cerebral artery occlusion (MCAO). Linagliptin was administered acutely (50 mg/kg intravenously), at MCAO time or chronically (10 mg/kg orally) for 4 weeks before and 3 weeks after MCAO. Neuroprotection was assessed by stroke volume measurement and quantification of NeuN-positive surviving neurons. Plasma/brain GLP-1 levels and dipeptidyl peptidase-4 activity were also measured. The results show that the linagliptin-mediated neuroprotection against stroke requires chronic pretreatment and does not occur via GLP-1R. The findings provide essential new knowledge with regard to the potential clinical use of gliptins against stroke, as well as a strong impetus to identify gliptin-mediated neuroprotective mechanisms.


Subject(s)
Cerebral Cortex/drug effects , Corpus Striatum/drug effects , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide 1/agonists , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Stroke/prevention & control , Administration, Oral , Animals , Cell Survival/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA-Binding Proteins , Dipeptidyl Peptidase 4/blood , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Exenatide , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Injections, Intravenous , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Nuclear Proteins/metabolism , Peptides/administration & dosage , Peptides/therapeutic use , Stroke/blood , Stroke/metabolism , Stroke/pathology , Venoms/administration & dosage , Venoms/therapeutic use
3.
J Neuroendocrinol ; 12(11): 1051-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11069120

ABSTRACT

We have recently identified nip-2 as a gene target for 17beta-oestradiol activity in the neuroblastoma SK-ER3 cells expressing the oestrogen receptor (ER) alpha. Here we show 17beta-oestradiol treatment of neuroblastoma and rat embryo neurones in culture blocks the increase in nip-2 mRNA induced by apoptotic stimuli and prevents cell death as indicated by cell counting, 3,(4,5-dimethylthiazol-2-yl)2,5-diphenil-tetrazoliumbromi de and DNA fragmentation assays. Neither of these effects are observed in the presence of the specific ER antagonist ICI 182,780, and are absent in neuroblastoma cells not expressing ER. We propose that nip-2 plays a relevant role in neural cell apoptosis and that a decrease in its expression is instrumental for the oestrogen anti-apoptotic effect described here. The experimental evidence presented supports the recent hypothesis of a protective role of oestrogens in neurodegenerative diseases such as Alzheimer's disease and highlights the importance of the development of new ER ligands for the prevention of neural cell damage.


Subject(s)
Apoptosis/drug effects , Calcium-Binding Proteins/genetics , Carrier Proteins , Estradiol/analogs & derivatives , Estradiol/pharmacology , Gene Expression/drug effects , Neurons/drug effects , Animals , Calcium-Binding Proteins/physiology , Cell Count , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , DNA Fragmentation , Embryo, Mammalian , Fulvestrant , Glucose/administration & dosage , Neuroblastoma/pathology , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2 , RNA, Messenger/analysis , Rats , Receptors, Estrogen/antagonists & inhibitors , Transfection , Tumor Cells, Cultured
4.
Endocrinology ; 141(5): 1839-45, 2000 May.
Article in English | MEDLINE | ID: mdl-10803594

ABSTRACT

Estrogens are female sex steroids that have a plethora of effects on a wide range of tissues. These effects are mediated through two well characterized intracellular receptors: estrogen receptor alpha and beta (ERalpha and ERbeta, respectively). Because of their high structural homology, it has been argued whether these two receptors may elicit differential biochemical events in estrogen target cells. Here we examine the effect of 17beta-estradiol-dependent activation of ERalpha and ERbeta on neurite sprouting, a well known consequence of this sex hormone action in neural cells. In SK-N-BE neuroblastoma cells transfected with ERalpha or ERbeta, 17beta-estradiol induces two distinct morphological phenotypes. ERalpha activation results in increased length and number of neurites, whereas ERbeta activation modulates only neurite elongation. By the use of chimeric receptors we demonstrate that the presence of both transcription activation functions located in the NH2-terminus and COOH-terminus of the two ER proteins are necessary for maintaining the differential biological activity reported. ERalpha-dependent, but not ERbeta-dependent, morphological changes are observed only in the presence of the active form of the small G protein Rac1B. Our data provide the first clear evidence that, in a given target cell, ERalpha and ERbeta may play distinct biological roles and support the hypothesis that 17beta-estradiol activates selected intracellular signaling pathways depending on the receptor subtype bound.


Subject(s)
Estradiol/pharmacology , Neurons/drug effects , Receptors, Estrogen/physiology , Animals , Cells, Cultured , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Neurites/metabolism , Neurons/metabolism , Phenotype , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Transfection
5.
Proc Natl Acad Sci U S A ; 96(19): 10905-10, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10485924

ABSTRACT

Estrogen is known to influence different functions in brain tissue ranging from neuronal development to plasticity and survival, but the mechanisms involved have not been defined clearly. Previous studies have shown the presence of the two estrogen receptors (ERs), ERalpha and ERbeta, in several brain areas, but less is known about the role of estrogen in the peripheral nervous system. Here we demonstrate that dorsal root ganglion (DRG) neurons express ERalpha and ERbeta during early postnatal development and in culture, and that the ERs localize mainly to neuronal cell nuclei. Studying the role of estrogen in DRG, we observed that low concentrations of 17beta-estradiol increased survival of cultured DRG neurons deprived of nerve growth factor. 17beta-Estradiol up-regulated the expression of the antiapoptotic molecule Bcl-x without affecting that of Bax, suggesting a mechanism by which the hormone counteracted neuronal death. Antiestrogens abolished the action of 17beta-estradiol in the DRG neurons, which demonstrates an involvement of ERs. The results show that estrogen and ERs play an important role in the development and survival of DRG neurons.


Subject(s)
Ganglia, Spinal/embryology , Gene Expression Regulation, Developmental , Neurons, Afferent/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Estradiol/pharmacology , Estradiol/physiology , Estrogen Receptor alpha , Estrogen Receptor beta , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Time Factors , Up-Regulation , bcl-2-Associated X Protein , bcl-X Protein
6.
Neurosci Lett ; 265(3): 207-10, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10327167

ABSTRACT

The neuropeptide pituitary adenylate cyclase activating polypeptide (PACAP) is expressed in various parts of the developing and adult rat brain, including the cerebellum. In situ hybridization was employed to localize the precise site of mRNA expression for PACAP and PACAP receptor I (PRI). During prenatal cerebellar development, PACAP mRNA was present in developing Purkinje cells and some deep cerebellar nuclei, whilst PRI mRNA was expressed by adjacent cells in the Purkinje cell layer (PCL). There was a shift in PRI mRNA expression to the external germinal cell layer around birth. PACAP decreased the number of neurons positive for the inhibitory neurotransmitter gamma-amino butyric acid (GABA) in cultures from embryonic cerebellum, but did not affect overall cell survival. In conclusion, our results show the pattern of PACAP mRNA expression in embryonic cerebellum and suggest a physiological role for PACAP on GABAergic cerebellar neurons.


Subject(s)
Neuropeptides/genetics , Neurotransmitter Agents/genetics , Pituitary Gland/chemistry , Purkinje Cells/physiology , gamma-Aminobutyric Acid/analysis , Age Factors , Animals , Cells, Cultured , Cerebellum/chemistry , Cerebellum/cytology , Cerebellum/embryology , Fetus/cytology , Gene Expression Regulation, Developmental , In Situ Hybridization , Medulla Oblongata/chemistry , Medulla Oblongata/embryology , Pituitary Adenylate Cyclase-Activating Polypeptide , Purkinje Cells/chemistry , Purkinje Cells/cytology , RNA, Messenger/analysis , Rats , Rats, Wistar
7.
Mol Endocrinol ; 12(6): 835-41, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626659

ABSTRACT

The alpha-estrogen receptor (ER alpha) transcriptional activity can be regulated either by binding to the cognate ligand or by intracellular signaling pathways responsive to a variety of factors acting through cell membrane receptors. Studies carried out in HeLa and COS-1 cells demonstrated that the cross-coupling between estrogen and growth factor receptors is mediated by p21ras and requires phosphorylation of a specific serine residue (Ser 118 in the human ER alpha and Ser 122 in mouse ER alpha) located in the ER alpha N-terminal activation function 1 (AF-1). Likewise, in the SK-N-BE neuroblastoma cell line p21ras is involved in the cross-coupling between insulin and ER alpha receptors. However, in this cell line Ser 122 is not necessary for insulin-dependent activation of unliganded ER alpha. In addition, after insulin activation, the electrophoretic mobility associated to serine hyperphosphorylation of ER alpha in SK-N-BE and in COS-1 cells is different. Our study rules out the possibility of tyrosine phosphorylation in unliganded ER alpha activation by means of transactivation studies of ER alpha tyrosine mutants and analysis of Tyr phosphorylation immunoreactivity. The two cofactors for steroid receptors RIP 140 and SRC-1 do not seem to be specifically involved in the insulin-induced ER alpha transactivation. The present study demonstrates the possibility of an alternative, cell-specific pathway of cross-coupling between intracellular and membrane receptors, which might be of importance for the understanding of the physiological significance of this mode of activation in the nervous system.


Subject(s)
Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic/physiology , Kidney Neoplasms/pathology , Neoplasm Proteins/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins p21(ras)/physiology , Receptor, IGF Type 1/physiology , Receptor, Insulin/physiology , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Animals , Estrogen Receptor alpha , HeLa Cells , Humans , Insulin/pharmacology , Ligands , Mice , Phosphorylation , Phosphoserine/chemistry , Protein Processing, Post-Translational , Receptor, IGF Type 1/drug effects , Receptor, Insulin/drug effects , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured
8.
Mol Endocrinol ; 11(7): 938-49, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9178753

ABSTRACT

Antisense oligos complementary to the 5'-end, but not to the 3'-end, of the estrogen receptor (ER) messenger RNA caused a paradox accumulation of ER protein in MCF-7 cells. The same effect was observed after treatment of the cells with the corresponding sense oligos. The oligos interfering with ER down-regulation were demonstrated to specifically bind the ER with affinities in the nanomolar range. It is, therefore, proposed that the ER up-regulation induced by the oligos might be due to squelching of the ER (or ER-inducible proteins) from their binding site located in the 5'-end of the ER gene. We also report that transcriptionally inactive ER mutants can undergo down-regulation, and that in denaturing gels, the migration profile of ER-oligo and ER-estrogen-responsive element complexes are dissimilar. We, therefore, propose that ER can interact with DNA in different ways and at different binding sites. These observations might have important pharmacological consequences, since specific drugs could be devised to induce the ER conformation necessary to perform only selected tasks of the ER transcriptional repertoire.


Subject(s)
Down-Regulation/genetics , Oligonucleotides, Antisense/genetics , Receptors, Estrogen/biosynthesis , Transfection/genetics , Animals , Base Sequence , Blotting, Western , COS Cells , DNA Primers/chemistry , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Humans , Luciferases/genetics , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Phosphorus Radioisotopes , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription, Genetic/physiology , Tumor Cells, Cultured , Urea/chemistry
9.
Eur J Neurosci ; 9(5): 1008-16, 1997 May.
Article in English | MEDLINE | ID: mdl-9182953

ABSTRACT

Oestrogens are the key factor in the sexual differentiation of the mammalian brain and play an important role in the activity of selected areas of the mature brain. To pursue the study of oestrogen action on neural cells at the molecular level, we developed a human neuroblastoma cell line (SK-ER3) expressing the oestrogen receptor (ER). Treatment of these cells with 17beta-oestradiol causes growth arrest and morphological and biochemical differentiation. The aim of the present study was to investigate whether oestrogen-differentiated SK-ER3 neuroblastoma cells acquire the ability to synthesize a specific neurotransmitter and whether the growth arrest previously reported can be ascribed to the blockage of the cells at a specific stage of the cell cycle. The results presented here indicate that oestrogens induce accumulation of SK-ER3 cells in the G0 phase of the cell cycle, underscoring the acquisition of a mature neural phenotype upon hormonal treatment. Most importantly, we show that in the differentiated cells the content of tyrosine hydroxylase and Na+-dependent dopamine uptake is significantly augmented, proving that the oestrogen-differentiated SK-ER3 cells can synthesize and store a specific neurotransmitter. In addition, we prove that the dopamine accumulated in differentiated SK-ER3 cells can be released. These studies therefore suggest that oestrogen treatment results in the acquisition of a fully functional dopaminergic phenotype of SK-ER3 cells. Ample evidence shows a link between dopaminergic neurons and oestrogen activity in hypothalamic and non-hypothalamic areas of the mammalian brain. Our study indicates that oestrogens might play a primary role in committing undifferentiated neuroblasts towards the dopaminergic phenotype.


Subject(s)
Dopamine/metabolism , Estrogens/pharmacology , Neuroblastoma/genetics , Neurons/drug effects , Humans , Immunohistochemistry , Phenotype , Tumor Cells, Cultured
10.
Brain Res Bull ; 44(4): 519-23, 1997.
Article in English | MEDLINE | ID: mdl-9370219

ABSTRACT

The neuroblastoma SK-ER3 cell line obtained by stable transfection of the human SK-N-BE cell line is proposed as a model for the study of estrogen receptor activity in cells of neural origin. In the SK-ER3 cell line the estrogen receptor, once activated, initiates a differentiation program leading to growth arrest, morphological changes, and acquisition of the dopaminergic phenotype. In the absence of estrogens, this program can be triggered by IGF-I, which can activate the unliganded estrogen receptor via the ras-pathway. It is proposed that this model system might recapitulate the events occurring in vivo during the differentiation of the nervous system and that IGF-I may play an important role in the activation of estrogen receptor at the very early stage of brain development affecting the differentiation of a number of hypothalamic and extrahypothalamic brain regions.


Subject(s)
Estradiol/pharmacology , Neurons/physiology , Receptors, Estrogen/physiology , Brain/physiology , Cell Differentiation , Cell Division/drug effects , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Kinetics , Male , Models, Neurological , Nerve Growth Factors/pharmacology , Neuroblastoma , Neurons/cytology , Neurons/drug effects , Receptor, IGF Type 1/physiology , Receptors, Estrogen/biosynthesis , Recombinant Proteins/biosynthesis , Sex Differentiation , Transfection , Tumor Cells, Cultured , Y Chromosome , ras Proteins/metabolism
11.
Mol Endocrinol ; 10(5): 499-507, 1996 May.
Article in English | MEDLINE | ID: mdl-8732681

ABSTRACT

Insulin is a well known mitotic agent for neuroblastoma cells. Human SK-N-BE neuroblastoma cells stably transfected with the estrogen receptor, however, undergo growth arrest and differentiation when treated with insulin. These effects were shown to be due to an insulin-dependent activation of the unliganded estrogen receptor. Here, we demonstrate that this activation involves the AF-2 COOH-terminal domain of the estrogen receptor and that the communication between estrogen and insulin receptor systems occurs via selected and specific transduction signals. In fact, by the use of dominant negative and dominant positive mutants we demonstrate that p21ras is essential for insulin and estrogen receptor coupling. With pharmacological tools, we prove that PI 3'kinase does not contribute to this cross-talk and that protein kinase C triggers transduction signals that act in synergism with p21ras. These results prove the intricacy of all these intracellular paths of communication. The finding that, in neuroblastoma cells, selected signal transduction systems are involved in the insulin-dependent activation of estrogen receptor is of particular interest considering that estrogen receptor might restrict the role played by insulin during the differentiation of neural cells and interfere with its proliferative potential while allowing its regulation of other functions related to cell survival.


Subject(s)
Insulin/pharmacology , Neuroblastoma/metabolism , Receptor, Insulin/physiology , Receptors, Estrogen/physiology , Cell Differentiation/drug effects , Cell Division/drug effects , Enzyme Activation , Genes, ras/physiology , Humans , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Receptors, Estrogen/genetics , Signal Transduction , Transcription, Genetic/drug effects , Transfection , Tumor Cells, Cultured
12.
DNA Cell Biol ; 14(8): 665-71, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7646813

ABSTRACT

In this work, we have studied the activity of a tetracycline modulatable trans-activator (tTA) generated by fusing the DNA binding domain of the tetracycline repressor to the trans-activation domain of the Herpes simplex virus protein 16 (HSV VP16) (plasmid pUHD15-1Neo). In the three different cell lines studied (HTC, rat hepatoma; T47D, human breast cancer; SK-N-BE, human neuroblastoma), the expression of the luciferase gene under the control of a tetracycline operator sequence (plasmid pUHC13-3) was used as a control of the incorporation and the functionality of the trans-activator. Clones selected from these cells responded in a time and dose-dependent manner to the withdrawal of tetracycline. In all these clones, the tTA trans-activator not only modulates the activity of the luciferase gene, but also modulates the activity of a number of endogenous proteins, including C/EBP beta, the glucocorticoid receptor (GR), and SP1. In the transfected cells, the level of these transcription factors was strongly inhibited in the presence of tetracycline and was highly increased after tetracycline removal. Electrophoresis mobility shift assay (EMSA) and footprint experiments proved that the induced proteins are perfectly efficient in binding the DNA. Their transcriptional activity was also determined. In HTC/A9 cells, the level of the chloramphenicol acetyltransferase (CAT) expression driven by the promoter of the alpha 1-glycoprotein (AGP) gene was strongly enhanced at 72-84 hr following removal of tetracycline from the growth media. The accumulation of the endogenous AGP mRNA also increased at 84 hr. In the T47D/TA11 and SK-N-BE/C2.6 cells, a general activation of protein synthesis was also evidenced.


Subject(s)
Genetic Vectors/genetics , Herpes Simplex Virus Protein Vmw65/metabolism , Repressor Proteins/metabolism , Tetracycline/pharmacology , Trans-Activators/metabolism , Animals , Base Sequence , Cytomegalovirus/genetics , DNA/metabolism , Gene Expression Regulation/drug effects , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Luciferases/biosynthesis , Luciferases/genetics , Molecular Sequence Data , Operator Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Rats , Receptors, Estradiol/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
13.
Mol Endocrinol ; 8(7): 910-8, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7984152

ABSTRACT

The neuroblastoma cell line SK-ER3, which is stably transfected with the estrogen receptor (ER), was used to study the effect of insulin and insulin-like growth factors (IGF-I and IGF-II) on growth and morphological differentiation induced by estrogens. The data demonstrate that insulin and related growth factors control the growth and morphological differentiation of the cell line expressing the ER, but not of the parental cell line. Effects elicited by the growth factors in SK-ER3 cells can be blocked by ER antagonists. Transient transfection studies further confirm an effect of the IGFs in modulation of ER-activated promoters. The results presented support the hypothesis of the existence of cross-talk between membrane and intracellular receptors and provide evidence for physiological consequences of the activation of such a pathway of communication. The present study is of particular interest with regard to the theory of prenatal involvement of the ER in maturation of nerve cells. It could, in fact, be hypothesized that IGF-I and IGF-II, present in high concentrations in the developing brain, might activate the ER expressed in several embryonic brain nuclei.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Insulin-Like Growth Factor II/pharmacology , Insulin-Like Growth Factor I/pharmacology , Neuroblastoma/pathology , Neurons/drug effects , Receptors, Estrogen/drug effects , Animals , Brain/embryology , Brain/physiology , Cell Differentiation/drug effects , Cell Division/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogens/physiology , Fulvestrant , Humans , Models, Neurological , Morphogenesis/physiology , Neoplasm Proteins/physiology , Pyrrolidines/pharmacology , Rats , Receptors, Estrogen/physiology , Recombinant Fusion Proteins/metabolism , Thiophenes/pharmacology , Transfection , Tumor Cells, Cultured/drug effects
14.
J Steroid Biochem Mol Biol ; 47(1-6): 207-11, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7903862

ABSTRACT

In order to assess the neuronal-like properties of a human neuroblastoma cell line obtained by stable transfection of the estrogen receptor (SK-ER3) a series of quantitative measurements of the activity of two neurotransmitter-related enzymes: tyrosine hydroxylase (TH) and monamine oxidase (MAO), and of catecholamine concentrations were performed. When compared to the parental SK-N-BE cell line, the stably transfected SK-ER3 cells show a more pronounced dopaminergic phenotype. The immunoreactivity to a TH antibody is in fact increased and the ratio between dopamine and noradrenaline concentrations is elevated. Treatment with estradiol further enhances the expression of this phenotype. Interestingly, in the transfected cell line MAO-A activity is decreased and further reduced by estrogen treatment. This finding substantiated by previous reports indicates that our model system might represent an interesting tool for the study of the pharmacological treatments of estrogen-induced pathological responses of nervous cells.


Subject(s)
Estradiol/pharmacology , Monoamine Oxidase/metabolism , Neuroblastoma/metabolism , Tyrosine 3-Monooxygenase/metabolism , Bucladesine/pharmacology , Dopamine/biosynthesis , Humans , Immunohistochemistry , Norepinephrine/biosynthesis , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL