Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Appl Opt ; 53(34): 8078-85, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25607966

RESUMO

The Atomic Energy Commission has set up a laboratory dedicated to the calibration of x-ray cameras mainly used in the Laser MégaJoule Facility in the Bordeaux region of France. Thanks to a double crystal monochromator specifically designed to perform such calibration, we calibrated a thinned, back-illuminated x-ray CCD manufactured by Roper Scientific Inc. over the 2.2-8.9 keV spectral region. Quantum efficiency results are in good agreement with those of previous works. To explain slight differences, we studied the difference between monochromatization methods performed either using a crystal monochromator or filters. Results showed that the latter method appears to be too dependent on the x-ray source parameters to perform accurate and relevant calibration on x-ray cameras.

2.
Gene Ther ; 20(3): 283-97, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22592599

RESUMO

Glioblastomas are the most aggressive of the brain tumors occurring in adults and children. Currently available chemotherapy prolongs the median survival time of patients by only 4 months. The low efficiency of current treatments is partly owing to the blood-brain barrier, which restricts the penetration of most drugs into the central nervous system. Locoregional treatment strategies thus become mandatory. In this context, viral tools are of great interest for the selective delivery of genes into tumoral cells. Gliomas express high levels of type 2 somatostatin receptors (sstr2A), pinpointing them as suitable targets for the improvement of transduction efficiency in these tumors. We designed a new adenoviral vector based on the introduction of the full-length somatostatin (SRIF (somatotropin release-inhibiting factor)) sequence into the HI loop of the HAdV fiber protein. We demonstrate that (i) HAdV-5-SRIF uptake into cells is mediated by sstr2A, (ii) our vector drives high levels of gene expression in cells expressing endogenous sstr2A, with up to 65-fold enhancement and (iii) low doses of HAdV-5-SRIF are sufficient to infect high-grade human primary glioblastoma cells. Adenoviral vectors targeting SRIF receptors might thus represent a promising therapeutic approach to brain tumors.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Receptores de Somatostatina/genética , Transdução Genética/métodos , Adenoviridae/metabolismo , Sequência de Aminoácidos , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Células CHO , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Cricetinae , Cricetulus , Endocitose , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/farmacocinética , Glioblastoma/patologia , Glioblastoma/terapia , Células HEK293 , Humanos , Immunoblotting , Integrinas/metabolismo , Microscopia Confocal , Dados de Sequência Molecular , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Células Tumorais Cultivadas
3.
Domest Anim Endocrinol ; 72: 106438, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32388344

RESUMO

The blood-brain barrier regulates the transport of molecules that convey global energetic status to the feeding circuitry within the hypothalamus. Capillaries within the median eminence (ME) and tight junctions between tanycytes lining the third ventricle (3V) are critical components of this barrier. Herein, we tested the hypothesis that altering the plane of nutrition results in the structural reorganization of tanycytes, tight junctions, and capillary structure within the medial basal hypothalamus. Proopiomelanocortin (POMC) neuronal content within the arcuate nucleus of the hypothalamus (ARC) was also assessed to test whether reduced nutritional status improved access of nutrients to the ARC, while decreasing the access of nutrients of overfed animals. Multiparous, nongestating ewes were stratified by weight and randomly assigned to dietary treatments offered for 75 d: 200% of dietary recommendations (overfed), 100% of dietary recommendations (control), or 60% of dietary recommendations (underfed). The number of POMC-expressing neurons within the ARC was increased (P ≤ 0.002) in underfed ewes. Overfeeding increased (P ≤ 0.01) tanycyte cellular process penetration and density compared with control and underfeeding as assessed using vimentin immunostaining. Immunostaining of tight junctions along the wall of the 3V did not differ (P = 0.32) between treatments. No differences were observed in capillary density (P = 0.21) or classification (P ≥ 0.47) within the ME. These results implicate that changes within the satiety center and morphology of tanycytes within the ARC occur as an adaptation to nutrient availability.


Assuntos
Células Ependimogliais/fisiologia , Hipotálamo/citologia , Plasticidade Neuronal/fisiologia , Ovinos , Ração Animal , Fenômenos Fisiológicos da Nutrição Animal , Animais , Adesão Celular , Dieta/veterinária , Metabolismo Energético , Feminino , Regulação da Expressão Gênica , Homeostase , Neurônios/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
4.
Oncogene ; 26(19): 2695-706, 2007 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-17057735

RESUMO

An instability of the mature cell phenotype is thought to participate to the formation of gliomas, primary brain tumors deriving from astrocytes and/or neural stem cells. Transforming growth factor alpha (TGFalpha) is an erbB1 ligand overexpressed in the earliest stages of gliomas, and exerts trophic effects on gliomal cells and astrocytes. Here, we questioned whether prolonged TGFalpha exposure affects the stability of the normal mature astrocyte phenotype. We first developed astrocyte cultures devoid of residual neural stem cells or progenitors. We demonstrate that days of TGFalpha treatment result in the functional conversion of a population of mature astrocytes into radial glial cells, a population of neural progenitors. TGFalpha-generated radial glial cells support embryonic neurons migration, and give birth to cells of the neuronal lineage, expressing neuronal markers and the electrophysiological properties of neuroblasts. Lengthening TGFalpha treatment to months results in the delayed appearance of cells with neural stem cells properties: they form floating cellular spheres that are self-renewing, can be clonally derived from a single cell and differentiated into cells of the neuronal lineage. This study uncovers a novel population of mature astrocytes capable, in response to a single epigenetic factor, to regress progressively into a neural stem-like cell stage via an intermediate progenitor stage.


Assuntos
Astrócitos/citologia , Diferenciação Celular/efeitos dos fármacos , Neurônios/metabolismo , Células-Tronco/citologia , Fator de Crescimento Transformador alfa/farmacologia , Animais , Astrócitos/metabolismo , Linhagem da Célula , Movimento Celular , Células Cultivadas , Eletrofisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Receptores ErbB/metabolismo , Feminino , Feto/citologia , Feto/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/citologia , Neurônios/citologia , Receptor ErbB-2/metabolismo , Proteínas Recombinantes/farmacologia , Células-Tronco/metabolismo
5.
Oncogene ; 25(29): 4076-85, 2006 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-16532035

RESUMO

Astrocyte death has been implicated in several neuropathological diseases, but the identification of molecules susceptible of promoting astrocyte survival has been elusive. We investigated whether transforming growth factor alpha (TGFalpha), an erbB1/EGFR ligand, which promotes glioma progression and affects astrocyte metabolism at embryonic and adult stages, regulates astrocyte survival. Primary serum-free astrocyte cultures from post-natal mouse and fetal human cortices were used. Transforming growth factor alpha protected both species of astrocytes from staurosporine-induced apoptosis. In serum-free medium, mouse astrocytes did not survive beyond 2 months while TGFalpha-treated astrocytes survived up to 12 months. Transforming growth factor alpha also promoted long-term survival of human astrocytes. We additionally extended TGFalpha proliferative effects to human astrocytes. After 3 days of permanent application, TGFalpha induced a major downregulation of both erbB1 and erbB2. This downregulation did not impair the functional activation of the receptors, as ascertained by their tyrosine phosphorylation and the continuous stimulation of both ERK/MAPK and PI3K/Akt pathways up to 7 days, the longest time examined. The full cellular effects of TGFalpha required activation of both transduction pathways. Enhanced proliferation and survival thus define TGFalpha as a gliatrophin for mammalian astrocytes. These results demonstrate that in normal, non-transformed astrocytes, sustained and functional erbBs activation is achieved without bypassing ligand-induced receptors downregulation.


Assuntos
Astrócitos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Envelhecimento/metabolismo , Animais , Astrócitos/citologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebelar/citologia , Córtex Cerebelar/embriologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioma/metabolismo , Humanos , Camundongos , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador alfa/metabolismo
6.
J Neuroendocrinol ; 19(9): 691-702, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17680884

RESUMO

In rodents, there is compelling evidence indicating that dynamic cell-to-cell communications involving cross talk between astroglial cells (such as astrocytes and specialised ependymoglial cells known as tanycytes) and neurones are important in regulating the secretion of gonadotrophin-releasing hormone (GnRH), the neurohormone that controls both sexual maturation and adult reproductive function. However, whether such astroglial cell-GnRH neurone interactions occur in the human brain is not known. In the present study, we used immunofluorescence to examine the anatomical relationship between GnRH neurones and glial cells within the hypothalamus of five women. Double-staining experiments demonstrated the ensheathment of GnRH neurone perikarya by glial fibrillary acidic protein (GFAP)-immunoreactive astrocyte processes in the periventricular zone of the tuberal region of the hypothalamus. GFAP immunoreactivity did not overlap that of GnRH at the GnRH neurone's projection site (i.e. the median eminence of the hypothalamus). Rather, human GnRH neuroendocrine fibres were found to be closely associated with vimentin or nestin-immunopositive radial glial processes likely belonging to tanycytes. In line with these light microscopy data, ultrastructural examination of GnRH-immunoreactive neurones showed numerous glial cells in direct apposition to pre-embedding-labelled GnRH cell bodies and/or dendrites in the infundibular nucleus, whereas postembedding immunogold-labelled GnRH nerve terminals were often seen to be enwrapped by glial cell processes in the median eminence. GnRH nerve button were sometimes visualised in close proximity to fenestrated pituitary portal blood capillaries and/or evaginations of the basal lamina that delineate the pericapillary space. In summary, these data demonstrate that GnRH neurones morphologically interact with astrocytes and tanycytes in the human brain and provide evidence that glial cells may contribute physiologically to the process by which the neuroendocrine brain controls the function of GnRH neurones in humans.


Assuntos
Astrócitos , Hormônio Liberador de Gonadotropina/análise , Hipotálamo , Neurônios , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Astrócitos/química , Astrócitos/citologia , Forma Celular , Feminino , Proteína Glial Fibrilar Ácida/análise , Humanos , Hipotálamo/anatomia & histologia , Hipotálamo/química , Proteínas de Filamentos Intermediários/análise , Proteínas do Tecido Nervoso/análise , Nestina , Plasticidade Neuronal , Neurônios/química , Neurônios/citologia , Vimentina/análise
7.
J Hazard Mater ; 325: 136-156, 2017 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-27930998

RESUMO

Triketones, derived chemically from a natural phytotoxin (leptospermone), are a good example of allelochemicals as lead molecules for the development of new herbicides. Targeting a new and key enzyme involved in carotenoid biosynthesis, these latest-generation herbicides (sulcotrione, mesotrione and tembotrione) were designed to be eco-friendly and commercialized fifteen-twenty years ago. The mechanisms controlling their fate in different ecological niches as well as their toxicity and impact on different organisms or ecosystems are still under investigation. This review combines an overview of the results published in the literature on ß-triketones and more specifically, on the commercially-available herbicides and includes new results obtained in our interdisciplinary study aiming to understand all the processes involved (i) in their transfer from the soil to the connected aquatic compartments, (ii) in their transformation by photochemical and biological mechanisms but also to evaluate (iii) the impacts of the parent molecules and their transformation products on various target and non-target organisms (aquatic microorganisms, plants, soil microbial communities). Analysis of all the data on the fate and impact of these molecules, used pure, as formulation or in cocktails, give an overall guide for the assessment of their environmental risks.


Assuntos
Herbicidas/análise , Herbicidas/química , Cetonas/análise , Cetonas/química , Cicloexanonas/análise , Ecossistema , Ecotoxicologia , Meio Ambiente , Concentração de Íons de Hidrogênio , Mesilatos/análise , Fotoquímica , Plantas/efeitos dos fármacos , Medição de Risco , Solo , Microbiologia do Solo , Sulfonas/análise , Temperatura , Água , Poluentes Químicos da Água/química
8.
Rev Sci Instrum ; 87(3): 033706, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27036783

RESUMO

The Laser Megajoule (LMJ) facility located at CEA/CESTA started to operate in the early 2014 with two quadruplets (20 kJ at 351 nm) focused on target for the first experimental campaign. We present here the first set of gated x-ray imaging (GXI) diagnostics implemented on LMJ since mid-2014. This set consists of two imaging diagnostics with spatial, temporal, and broadband spectral resolution. These diagnostics will give basic measurements, during the entire life of the facility, such as position, structure, and balance of beams, but they will also be used to characterize gas filled target implosion symmetry and timing, to study x-ray radiography and hydrodynamic instabilities. The design requires a vulnerability approach, because components will operate in a harsh environment induced by neutron fluxes, gamma rays, debris, and shrapnel. Grazing incidence x-ray microscopes are fielded as far as possible away from the target to minimize potential damage and signal noise due to these sources. These imaging diagnostics incorporate microscopes with large source-to-optic distance and large size gated microchannel plate detectors. Microscopes include optics with grazing incidence mirrors, pinholes, and refractive lenses. Spatial, temporal, and spectral performances have been measured on x-ray tubes and UV lasers at CEA-DIF and at Physikalisch-Technische Bundesanstalt BESSY II synchrotron prior to be set on LMJ. GXI-1 and GXI-2 designs, metrology, and first experiments on LMJ are presented here.

9.
Circulation ; 101(13): 1594-7, 2000 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-10747354

RESUMO

BACKGROUND: Although estrogen replacement therapy has been associated with reduction of cardiovascular events in postmenopausal women, the mechanism for this benefit remains unclear. Because nitric oxide (NO) is considered an important endothelium-derived relaxing factor and may function to protect blood vessels against atherosclerotic development, we investigated the acute effects of physiological levels of estrogen on NO release from human internal thoracic artery endothelia and human arterial endothelia in culture. METHODS AND RESULTS: We tested the hypothesis that estrogen acutely stimulates constitutive NO synthase activity in human endothelial cells by acting on a cell-surface receptor. NO release was measured in real time with an amperometric probe. 17beta-Estradiol exposure to internal thoracic artery endothelia and human arterial endothelia in culture stimulated NO release within seconds in a concentration-dependent manner. 17beta-Estradiol conjugated to bovine serum albumin also stimulated NO release, suggesting action through a cell-surface receptor. Tamoxifen, an estrogen receptor inhibitor, antagonized this action. We further showed with the use of dual emission microfluorometry that 17beta-estradiol-stimulated release of endothelial NO was dependent on the initial stimulation of intracellular calcium transients. CONCLUSIONS: Physiological doses of estrogen immediately stimulate NO release from human endothelial cells through activation of a cell-surface estrogen receptor that is coupled to increases in intracellular calcium.


Assuntos
Cálcio/fisiologia , Membrana Celular/metabolismo , Endotélio Vascular/metabolismo , Óxido Nítrico/metabolismo , Receptores de Estrogênio/fisiologia , Adulto , Idoso , Artérias/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Humanos , Técnicas In Vitro , Membranas Intracelulares/metabolismo , Masculino , Tamoxifeno/farmacologia
10.
Mol Cell Biol ; 35(14): 2368-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25939386

RESUMO

Amyloid ß (Aß) peptides originating from amyloid precursor protein (APP) in the endosomal-lysosomal compartments play a critical role in the development of Alzheimer's disease (AD), the most common type of senile dementia affecting the elderly. Since insulin-like growth factor II (IGF-II) receptors facilitate the delivery of nascent lysosomal enzymes from the trans-Golgi network to endosomes, we evaluated their role in APP metabolism and cell viability using mouse fibroblast MS cells deficient in the murine IGF-II receptor and corresponding MS9II cells overexpressing the human IGF-II receptors. Our results show that IGF-II receptor overexpression increases the protein levels of APP. This is accompanied by an increase of ß-site APP-cleaving enzyme 1 levels and an increase of ß- and γ-secretase enzyme activities, leading to enhanced Aß production. At the cellular level, IGF-II receptor overexpression causes localization of APP in perinuclear tubular structures, an increase of lipid raft components, and increased lipid raft partitioning of APP. Finally, MS9II cells are more susceptible to staurosporine-induced cytotoxicity, which can be attenuated by ß-secretase inhibitor. Together, these results highlight the potential contribution of IGF-II receptor to AD pathology not only by regulating expression/processing of APP but also by its role in cellular vulnerability.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Receptor IGF Tipo 2/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Endossomos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Microscopia de Fluorescência , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Interferência de RNA , Receptor IGF Tipo 2/genética , Estaurosporina/farmacologia , Transfecção
11.
Endocrinology ; 142(10): 4288-94, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11564686

RESUMO

Recent studies from our laboratory suggested that the vascular endothelium of the median eminence was involved via nitric oxide secretion in the modulation of GnRH release during the estrous cycle. To further investigate that issue, we studied the variations of endothelial nitric oxide synthase protein and mRNA in the median eminence of female rats killed at different time points of the day and/or of the estrous cycle. Endothelial nitric oxide synthase protein levels were measured by Western blot, and endothelial nitric oxide synthase mRNA analysis was performed with semiquantitative RT-PCR (for each time point, n = 4). The results revealed that endothelial nitric oxide synthase synthesis varied markedly across the estrous cycle. Indeed, endothelial nitric oxide synthase protein (n = 20) and mRNA (n = 16) levels increase significantly on 0800 h and 1600 h proestrus compared with 1400 h diestrus II. In a second step, quantification analysis were made in median eminence obtained from ovariectomized and ovariectomized, E2 benzoate primed rat. The results show a significant increase in expression of endothelial nitric oxide synthase protein as well as endothelial nitric oxide synthase mRNA in ovx-E2 primed rat median eminence. Concurrently, the levels of the cav-1 protein, a specific endogenous inhibitor of endothelial nitric oxide synthase, were measured in median eminence during estrous cycle and in ME from ovx and ovx-E2 primed rats. A significant decrease of median eminence cav-1 was noted on 1600 h proestrus and in ovx-E2 primed rats when compared with 1400 h diestrus II and ovx, respectively. Altogether, these results strongly suggest that high NO release from median eminence observed on proestrus may be due to an increase of endothelial nitric oxide synthase expression and a decrease of the cav-1 protein levels. These findings demonstrate that E2 is able to modulate endothelial nitric oxide synthase and cav-1 expression both during the estrous cycle and in experimental conditions and consequently reinforce the idea that nitric oxide acting on GnRH release, is essentially endothelial in origin. These results may also imply that variations of endothelial nitric oxide synthase expression are essential for the pulsatile/cyclic nitric oxide median eminence release observed in a previous study.


Assuntos
Estro/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Eminência Mediana/fisiologia , Óxido Nítrico Sintase/fisiologia , Animais , Vasos Sanguíneos/fisiologia , Endotélio Vascular/fisiologia , Feminino , Eminência Mediana/irrigação sanguínea , Óxido Nítrico Sintase Tipo III , Ratos , Ratos Wistar
12.
Endocrinology ; 142(6): 2343-50, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11356681

RESUMO

The involvement of nitric oxide (NO) as a gaseous neurotransmitter in the hypothalamic control of pituitary LH secretion has been demonstrated. NO, as a diffusible signaling gas, has the ability to control and synchronize the activity of the neighboring cells. NO is secreted at the median eminence (ME), the common termination field for the antehypophysiotropic neurons, under the stimulation of other signaling substances. At the ME, NO stimulates GnRH release from neuroendocrine terminals. The present studies were undertaken to determine whether NO is secreted spontaneously from ME fragments ex vivo and whether its secretion is correlated to GnRH release. To accomplish this, female rats were killed at different time points of the day and/or of the estrous cycle. The spontaneous NO release was monitored in real time, with an amperometric probe, during 4 periods of 30 min, from individual ME fragments (for each time point, n = 4). GnRH levels were measured in parallel for each incubation-period by RIA. The results revealed that NO was released in a pulsatile manner from female ME fragments and, unambiguously, that the amplitude of NO secretion varied markedly across the estrous cycle. Indeed, though the NO pulse period (32 +/- 1 min, n = 36) and duration (21 +/- 2 min, n = 36) did not vary significantly across the estrous cycle, the amplitude of this secretion pulse was significantly higher on proestrus (Pro; 39 +/- 3 nM, n = 20), compared with diestrus (16 +/- 1 nM, n = 8) or estrus (23 +/- 3 nM, n = 8, P < 0.05). The GnRH levels in the incubation medium were positively correlated to NO secretion across the estrous cycle (r = 0.86, P < 0.003, n = 9), confirming that NO and GnRH release are coupled. Furthermore, 5 x 10(-7) M L-N(5)-(1-iminoethyl)ornithine (L-NIO), a NO synthase inhibitor, succeeded in inhibiting the strong NO-GnRH secretory coupling and GnRH release on PRO: Because at this concentration, L-NIO selectively inhibits endothelial NO synthase, the results further demonstrate that the major source of NO involved in GnRH release at the ME is endothelial in origin. Additionally, the induction of a massive NO/GnRH release in 15-day ovariectomized rat treated with estradiol benzoate strongly suggested that estradiol is participating in the stimulation of NO release activity between diestrus II and PRO: The present study is the first demonstrating that ME can spontaneously release NO and that NO's rhythm of secretion varies markedly across the estrous cycle. This pulsatile/cyclic ME NO release may constitute the synchronizing link to anatomically scattered GnRH neurons.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/metabolismo , Óxido Nítrico/metabolismo , Animais , Inibidores Enzimáticos/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Estro , Feminino , Técnicas In Vitro , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III , Ornitina/análogos & derivados , Ornitina/farmacologia , Ovariectomia , Periodicidade , Ratos , Ratos Wistar
13.
Endocrinology ; 139(10): 4127-39, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9751492

RESUMO

Variations in the number of galanin receptor (Gal-R1)-expressing cells and levels of Gal-R1 messenger RNA (mRNA) were determined in the preoptic area in intact female rats throughout the phases of the estrous cycle and compared with those in the male. Female and male Wistar rats were fixed by perfusion with 4% paraformaldehyde. Cryostat sections were hybridized with a 35S-labeled antisense Gal-R1 riboprobe. The number of Gal-R1 mRNA-expressing cells was lower in the rostral preoptic area than in the medial preoptic area. During the estrous cycle, the highest number of Gal-R1 mRNA-expressing cells in the rostral preoptic region was detected at 0800 h on proestrus, whereas in the medial preoptic area, the maximum number was observed at 1800 h on estrus. Gal-R1 mRNA levels in individual cells were low during diestrus and increased at estrus in both areas. In the male, the number of mRNA-expressing cells and the hybridization signal were significantly lower than those in females during estrus. The results demonstrate that Gal-R1 gene expression in the preoptic area varies during the estrous cycle and is low in males. Short term treatment of ovariectomized rats with estradiol plus progesterone caused significantly decreased preoptic Gal-R1 mRNA levels compared with those after treatment with estrogen only. These observations suggest that in the preoptic area, expression of Gal-R1 is influenced by progesterone. The variation in Gal-R1 expression is likely to influence the extent to which galanin can influence the preoptic cells implicated in the control of neighboring GnRH cells.


Assuntos
Estro , Área Pré-Óptica/metabolismo , RNA Mensageiro/análise , Receptores de Neuropeptídeos/genética , Animais , Ritmo Circadiano , Feminino , Hormônio Luteinizante/sangue , Masculino , Ratos , Ratos Wistar , Receptores de Galanina , Fatores Sexuais
14.
Endocrinology ; 142(9): 4055-65, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11517185

RESUMO

The purpose of the present study was to determine whether TGF beta, a cytokine secreted by hypothalamic astrocytes, was able to regulate POMC neurons in the arcuate nucleus. In a first set of experiments, mediobasal hypothalamic fragments were exposed to TGF beta(1), and the relative POMC mRNA expression was assessed by in situ hybridization using a radiolabeled POMC riboprobe. The results showed that 4 x 10(-10) M TGF beta(1) was efficient in decreasing significantly the amounts of POMC mRNA (P < 0.01). Interestingly, the decrease of relative POMC mRNA levels was higher in the rostral than in the caudal parts of the arcuate nucleus. In a second set of experiments, we examined the occurrence of TGF beta receptors expression in arcuate POMC neurons. Dual labeling in situ hybridization and in situ hybridization, coupled to immunohistochemical labeling, were performed to examine mRNA expression of the type I serine-threonine kinase receptor for TGF beta and the presence of type II receptor for TGF beta, respectively, in POMC neurons. The results indicated that TGF beta receptor I mRNA and TGF beta receptor II protein were expressed in numerous POMC neurons. Regional analysis revealed that the highest proportion of POMC neurons expressing TGF beta receptors was located in the rostral part of the arcuate nucleus. Using dual labeling immunohistochemistry, we also found that Smad2/3 immunoreactivity, a TGF beta(1) downstream signaling molecule, was present in the cytoplasm and nucleus of some POMC (beta-endorphin) neurons. We next examined whether the number of POMC neurons expressing TGF beta-RI mRNA was affected by sex steroids. Quantification of the number of POMC neurons expressing TGF beta receptor I mRNA in ovariectomized, ovariectomized E2-treated, and ovariectomized E2 plus progesterone-treated animals revealed that estrogen treatment decreased the expression of TGF beta receptor I mRNA in POMC neurons located in the rostral half of the arcuate nucleus, an effect reversed by progesterone in a subset of the most rostral cells. Taken together, these data reveal that TGF beta(1) may directly modulate the activity of POMC neurons through the activation of TGF beta receptors. Therefore, the present study provides additional evidence for the involvement of TGF beta(1) in the regulation of neuroendocrine functions and supports the existence of a glial-to-neurons communication within the arcuate nucleus.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Pró-Opiomelanocortina/genética , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Sinergismo Farmacológico , Estradiol/farmacologia , Feminino , Hormônios/sangue , Ovariectomia , Progesterona/farmacologia , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/farmacologia
15.
Endocrinology ; 141(5): 1648-57, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10803573

RESUMO

We have shown previously at the ultrastructural level that morphological changes occur in the external zone of the median eminence allowing certain GnRH nerve terminals to contact the pericapillary space on the day of proestrus. The present study was designed to determine whether the intrinsic determinant of neuronal outgrowth, growth-associated protein-43 (GAP-43), was expressed in GnRH neurons of adult female rats, and whether its expression varied throughout the estrous cycle. To accomplish this, we perfusion-fixed groups of adult female rats at 0800 and 1600 h on diestrous day 2 (diestrous II), at 0800 h and 1600 h on proestrus, and at 0800 and 1600 h on estrus (n = 4 rats/group) and used double labeling in situ hybridization and quantification to compare the levels of GAP-43 messenger RNA (mRNA) in cells coexpressing GnRH mRNA. GnRH mRNA was detected with an antisense complementary RNA (cRNA) probe labeled with the hapten digoxigenin, whereas the GAP-43 cRNA probe was labeled with 35S and detected by autoradiography. In addition, GAP-43 protein was identified with immunohistochemistry in the median eminence. The results show that many GnRH neurons expressed GAP-43 mRNA and that GAP-43 protein was present in many GnRH axon terminals in the outer layer of the median eminence. The number of GnRH neurons expressing GAP-43 mRNA was significantly higher on proestrus (64 +/- 5%) than on diestrous II (40 +/- 2%; P < 0.001) or on estrus (45 +/- 8%; P < 0.05), and the GAP-43 mRNA levels in GnRH neurons also varied as a function of time of death during the estrous cycle. The GAP-43 mRNA levels in GnRH neurons were higher on proestrus and estrus than on diestrous II (P < 0.05). These data show that 1) GAP-43 is expressed in adult GnRH neurons; 2) GAP-43 mRNA expression in GnRH neurons fluctuates during the estrous cycle; and 3) GAP-43 mRNA content in GnRH neurons is highest on the day of proestrus, before and during the onset of the LH surge. These observations suggest that the increased GAP-43 mRNA expression in GnRH neurons on the day of proestrus could promote the outgrowth of GnRH axon terminals to establish direct neurovascular contacts in the external zone of the median eminence and thus facilitate GnRH release into the pituitary portal blood.


Assuntos
Proteína GAP-43/genética , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , RNA Mensageiro/biossíntese , Animais , Estrogênios/sangue , Estro , Feminino , Proteína GAP-43/biossíntese , Hormônio Luteinizante/sangue , Eminência Mediana/metabolismo , Terminações Pré-Sinápticas/metabolismo , Progesterona/sangue , Ratos , Ratos Wistar
16.
Endocrinology ; 141(5): 1780-94, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10803589

RESUMO

Previous studies have shown that galanin-containing fibers make synaptic contacts with POMC neurons in the arcuate nucleus. However, the ability of POMC neurons to express galanin receptors has never been assessed. The present study was designed to investigate whether POMC neurons express galanin receptor messenger RNA (mRNA) and whether testosterone could modulate galanin receptor gene expression. A dual-labeling in situ hybridization histochemistry, using 35S-labeled (galanin receptors GalR1 or GalR2) and digoxigenin-labeled (POMC) riboprobes, was performed on brain sections from intact, castrated, and testosterone-replaced adult male rats. For analysis, the arcuate nucleus was divided into four rostro-caudal areas. The results revealed that both GalR1 and GalR2 mRNAs were expressed in POMC neurons. Most POMC neurons expressing galanin receptor mRNAs were found in the rostral parts of the nucleus. Castration reduced the labeling density of galanin receptor mRNAs in POMC neurons, and testosterone prevented the effects of castration in all rostro-caudal subdivisions of the arcuate nucleus. Taken together, these data indicate that galanin can directly modulate the activity of POMC neurons, via an action on GalR1 or GalR2 receptors, particularly in the rostral-arcuate nucleus. In addition, testosterone can modulate the expression of GalR1 and GalR2. Because POMC neurons located in the rostral part of the nucleus are known to project preferentially to the preoptic area, POMC neurons expressing the galanin receptor genes may play an important role in the regulation of the GnRH neuroendocrine axis.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Neurônios/metabolismo , Pró-Opiomelanocortina , RNA Mensageiro/biossíntese , Receptores de Neuropeptídeos/genética , Testosterona/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Hibridização In Situ , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Galanina , Receptores de Neuropeptídeos/biossíntese
17.
Endocrinology ; 140(2): 652-9, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9927290

RESUMO

The median eminence (ME), which is the common termination field for adenohypophysiotropic systems, has been shown to produce nitric oxide (NO), a signaling molecule involved in neuroendocrine secretion. Using an ex vivo technique, 17beta-estradiol exposure to ME fragments, including vascular tissues, stimulated NO release within seconds in a concentration-dependent manner, whereas 17alpha-estradiol or testosterone had no effect. 17Beta-estradiol conjugated to BSA (E2-BSA) also stimulated NO release, suggesting mediation by a membrane surface receptor. Tamoxifen, an estrogen receptor inhibitor, antagonized the action of both 17beta-estradiol and E2-BSA. Furthermore, estradiol-stimulated NO stimulates GnRH release. This was demonstrated by hemoglobin (a NO scavenger), N(omega)-nitro-L-arginine methyl ester, and L-N5-(1-iminoethyl)ornithine (nitric oxide synthase inhibitors) inhibition of estradiol stimulated NO and GnRH release. In this regard, L-N5-(1-iminoethyl)ornithine, specific for endotheliol constitutive nitric oxide synthase, was significantly more potent, suggesting that the estradiol-stimulated NO release arose from vascular endothelial cells. Additionally, the NO-stimulated GnRH release occurs via guanylyl cyclase activation in GnRH nerve terminals, as ODQ, a potent and selective inhibitor of NO-sensitive guanylyl cyclase, abolished the estradiol-stimulated GnRH release. The results suggest that at physiological concentrations, 17beta-estradiol may have immediate actions on ME endothelial cells via nongenomic signaling pathways leading to NO-stimulated GnRH release.


Assuntos
Endotélio Vascular/metabolismo , Estradiol/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/metabolismo , Óxido Nítrico/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Combinação de Medicamentos , Ativação Enzimática/fisiologia , Estradiol/farmacologia , Estradiol/fisiologia , Guanilato Ciclase/metabolismo , Masculino , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/ultraestrutura , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Ratos , Ratos Wistar , Soroalbumina Bovina/farmacologia , Testosterona/farmacologia
18.
Neuroscience ; 84(1): 177-91, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9522372

RESUMO

The ultrastructural appearance of gonadotropin releasing hormone-immunoreactive elements was studied in the external zone of the median eminence of adult female Wistar rats. On the one hand, the purpose of the study was to determine the distribution of gonadotropin releasing hormone terminals towards the parenchymatous basal lamina at the level of hypothalamo-hypophyseal portal vessels, throughout the estrous cycle. On the other hand, we have semi-quantified the gonadotropin releasing hormone content in nerve terminals or preterminals during this physiological condition. A morphometric study was coupled to a colloidal 15 mn gold postembedding immunocytochemistry procedure. Animals were killed at 09.00 on diestrus II, 0.900, 10.00, 13.00, 17.00 and 18.00 on proestrus and 09.00 on estrus (n = 4-8 rats/group). A preliminary light microscopic study was carried out to identify an antero-posterior part of median eminence strongly immunostained by anti-gonadotropin releasing hormone antibodies but which was, in addition, easily spotted. This last condition was necessary to make a good comparison between each animal. Contacts between gonadotropin releasing hormone nerve terminals and the basal lamina were observed only the day of proestrus. Such contacts, however, were rare and in the great majority of cases, gonadotropin releasing hormone terminals are separated from basal lamina by tanycytic end feet. The morphometric analysis showed no significant variation in average distance between gonadotropin releasing hormone terminals and capillaries throughout the estrous cycle. Consequently, it did not appear that a large neuroglial plasticity exists during the estrous cycle. However, the observation of contacts only on proestrus together with some ultrastructural images evoke the possibility of a slight plasticity. The semi-quantitative results show that the content of gonadotropin releasing hormone in the nerve endings presented two peaks on proestrus: one at 09.00 (23 +/- 5 particles/micrograms2, P < 0.03) before the onset of luteinizing hormone surge, and the second at 18.00 (16 +/- 2 particles/micrograms2, P < 0.01) concomitantly with the luteinizing hormone surge, when compared to baseline values on proestrus 10.00 (8 +/- particles/micrograms2).


Assuntos
Estro/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/metabolismo , Terminações Nervosas/metabolismo , Neuropeptídeos/metabolismo , Animais , Feminino , Fase Folicular/metabolismo , Hormônios/metabolismo , Imuno-Histoquímica , Eminência Mediana/ultraestrutura , Microscopia Eletrônica , Ratos , Ratos Wistar , Distribuição Tecidual
19.
Neuroscience ; 94(3): 809-19, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10579572

RESUMO

Despite intense investigation, the demonstration of morphological plasticity in the external zone of the median eminence concerning the gonadotropin-releasing hormone system has never been reported. In this study, we investigate whether dynamic transformations of the gonadotropin-releasing hormone nerve terminals and/or tanycytes in the external zone of the median eminence of the hypothalamus occurred during the rat estrous cycle, by following individual gonadotropin-releasing hormone-immunoreactive nerve terminals on serial ultrathin sections observed by electron microscopy. Female rats were killed at 16.00 diestrus II (n = 3), i.e. when estrogen levels are basal and gonadotropin-releasing hormone release is low, and at 16.00 proestrus (n = 4), i.e. when estrogen levels peak and the preovulatory gonadotropin-releasing hormone surge occurs. Our results show that, in the median eminence obtained from proestrus rats, 12+/-2% of the gonadotropin-releasing hormone nerve terminals were observed to make physical contact with the parenchymatous basal lamina, i.e. the pericapillary space. In the median eminence obtained from diestrus II rats, no contacts were observed. On proestrus, numerous physical contacts between gonadotropin-releasing hormone nerve terminals and the basal lamina occurred by evagination of the basal lamina and/or by emerging processes from gonadotropin-releasing hormone nerve terminals. The quantification of the evagination of the basal lamina revealed that the basal lamina was at least twofold more tortuous in appearance during proestrus. These results demonstrate for the first time the existence of dynamic plastic changes in the external zone of the median eminence, allowing gonadotropin-releasing hormone nerve terminals to contact the pericapillary space on the day of proestrus, thus facilitating the release of the neurohormone into the pituitary portal blood.


Assuntos
Endotélio Vascular/fisiologia , Estro/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Eminência Mediana/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Animais , Diestro , Estrogênios/sangue , Feminino , Imuno-Histoquímica , Hormônio Luteinizante/sangue , Eminência Mediana/citologia , Terminações Nervosas/fisiologia , Terminações Nervosas/ultraestrutura , Neuroglia/citologia , Plasticidade Neuronal , Neurônios/citologia , Proestro , Ratos , Ratos Wistar
20.
Neuroscience ; 112(2): 475-85, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12044465

RESUMO

It has become apparent that galanin as well as proopiomelanocortin-derived peptides, such as beta-endorphin, play an important role in the hypothalamic circuitry that regulates neuroendocrine functions and appetite behavior. We have recently shown that GalR1 and GalR2 galanin receptor mRNAs are expressed in proopiomelanocortin neurons of the arcuate nucleus, suggesting a direct modulatory action of galanin on the proopiomelanocortin neuronal system. In the present study, we investigated the effect of galanin on beta-endorphin release and proopiomelanocortin mRNA expression from male rat mediobasal hypothalamic fragments incubated ex vivo. Galanin induced a decrease of spontaneous beta-endorphin release within the first 30-60 min of incubation and this effect was blocked by the galanin receptor antagonist galantide. Co-incubation of galanin with FK-506 (tacrolimus), a calcineurin inhibitor, suppressed the inhibitory effect of galanin on beta-endorphin release, suggesting that calcineurin is involved in the galanin-evoked decrease in beta-endorphin release. Measurement of beta-endorphin levels in the tissues at the end of the incubation period (120 min) revealed that galanin caused a two-fold increase of beta-endorphin peptide concentration in the mediobasal hypothalamic tissues. Concurrently, galanin induced an increase in the mean density of silver grains overlying proopiomelanocortin neurons after 60 min of incubation, an effect antagonized by galantide. Finally, reverse transcription-polymerase chain reaction analysis revealed that the mRNAs for the three galanin receptor subtypes (i.e. GalR1, GalR2, and GalR3) were expressed in the incubated mediobasal hypothalamic fragments. Taken as a whole, our results indicate that galanin plays a modulatory role on proopiomelanocortin neurons and this interrelation contributes to the elucidation of the neural circuitry that controls, among others, gonadotropin-releasing hormone function.


Assuntos
Galanina/farmacologia , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Pró-Opiomelanocortina/metabolismo , Animais , Inibidores de Calcineurina , Relação Dose-Resposta a Droga , Galanina/antagonistas & inibidores , Hipotálamo/química , Hipotálamo/citologia , Hipotálamo/fisiologia , Hibridização In Situ , Técnicas In Vitro , Masculino , Neurônios/química , Neurônios/citologia , Neurônios/fisiologia , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptores de Galanina , Receptores de Neuropeptídeos/biossíntese , Receptores de Neuropeptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tacrolimo/farmacologia , beta-Endorfina/análise , beta-Endorfina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA