Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Domest Anim Endocrinol ; 72: 106438, 2020 07.
Article in English | MEDLINE | ID: mdl-32388344

ABSTRACT

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.


Subject(s)
Ependymoglial Cells/physiology , Hypothalamus/cytology , Neuronal Plasticity/physiology , Sheep , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Cell Adhesion , Diet/veterinary , Energy Metabolism , Female , Gene Expression Regulation , Homeostasis , Neurons/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
2.
J Hazard Mater ; 325: 136-156, 2017 Mar 05.
Article in English | MEDLINE | ID: mdl-27930998

ABSTRACT

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.


Subject(s)
Herbicides/analysis , Herbicides/chemistry , Ketones/analysis , Ketones/chemistry , Cyclohexanones/analysis , Ecosystem , Ecotoxicology , Environment , Hydrogen-Ion Concentration , Mesylates/analysis , Photochemistry , Plants/drug effects , Risk Assessment , Soil , Soil Microbiology , Sulfones/analysis , Temperature , Water , Water Pollutants, Chemical/chemistry
3.
Rev Sci Instrum ; 87(3): 033706, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27036783

ABSTRACT

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.

4.
Mol Cell Biol ; 35(14): 2368-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939386

ABSTRACT

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.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Receptor, IGF Type 2/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Endosomes/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lysosomes/metabolism , Mice , Microscopy, Fluorescence , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , Receptor, IGF Type 2/genetics , Staurosporine/pharmacology , Transfection
5.
Appl Opt ; 53(34): 8078-85, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25607966

ABSTRACT

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.

6.
Gene Ther ; 20(3): 283-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22592599

ABSTRACT

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.


Subject(s)
Adenoviridae/genetics , Brain Neoplasms/genetics , Glioblastoma/genetics , Receptors, Somatostatin/genetics , Transduction, Genetic/methods , Adenoviridae/metabolism , Amino Acid Sequence , Animals , Brain Neoplasms/pathology , Brain Neoplasms/therapy , CHO Cells , Capsid Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Cricetinae , Cricetulus , Endocytosis , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/pharmacokinetics , Glioblastoma/pathology , Glioblastoma/therapy , HEK293 Cells , Humans , Immunoblotting , Integrins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Receptors, Somatostatin/metabolism , Somatostatin/genetics , Somatostatin/metabolism , Tumor Cells, Cultured
7.
J Neuroendocrinol ; 24(5): 798-808, 2012 May.
Article in English | MEDLINE | ID: mdl-22512326

ABSTRACT

GPR50, formerly known as melatonin-related receptor, is one of three subtypes of the melatonin receptor subfamily, together with the MT(1) and MT(2) receptors. By contrast to these two high-affinity receptor subtypes and despite its high identity with the melatonin receptor family, GPR50 does not bind melatonin or any other known ligand. Specific and reliable immunological tools are therefore needed to be able to elucidate the physiological functions of this orphan receptor that are still largely unknown. We have generated and validated a new specific GPR50 antibody against the ovine GPR50 and used it to analyse the neuroanatomical distribution of the GPR50 in sheep, rat and mouse whole brain. We demonstrated that GPR50-positive cells are widely distributed in various regions, including the hypothalamus and the pars tuberalis of the pituitary, in all the three species studied. GPR50 expressing cells are abundant in the dorsomedial nucleus of the hypothalamus, the periventricular nucleus and the median eminence. In rodents, immunohistochemical studies revealed a broader distribution pattern for the GPR50 protein. GPR50 immunoreactivity is found in the medial preoptic area (MPA), the lateral septum, the lateral hypothalamic area, the bed nucleus of the stria terminalis, the vascular organ of the laminae terminalis and several regions of the amygdala, including the medial nuclei of amygdala. Additionally, in the rat brain, GPR50 protein was localised in the CA1 pyramidal cell layer of the dorsal hippocampus. In mice, moderate to high numbers of GPR50-positive cells were also found in the subfornical organ. Taken together, these results provide an enlarged distribution of GPR50 protein, give further insight into the organisation of the melatoninergic system, and may lay the framework for future studies on the role of the GPR50 in the brain.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Rodentia/metabolism , Sheep/metabolism , Age Factors , Animals , Brain/anatomy & histology , Female , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Rabbits , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Rodentia/anatomy & histology , Rodentia/genetics , Sheep/anatomy & histology , Sheep/genetics , Tissue Distribution
8.
J Neuroendocrinol ; 22(7): 639-49, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20492366

ABSTRACT

Although coordinated actions of several areas within the hypothalamus are involved in the secretion of gonadotrophin-releasing hormone (GnRH), the median eminence of the hypothalamus, where the nerve terminals are located, plays a particularly critical role in the release of GnRH. In adult females, prior to the preovulatory surge of GnRH, the retraction of specialised ependymoglial cells lining the floor of the third ventricle named tanycytes allows for the juxtaposition of GnRH nerve terminals with the adjacent pericapillary space of the pituitary portal vasculature, thus forming direct neurohaemal junctions. These morphological changes occur within a few hours and are reversible. Such remodelling may promote physiological conditions to enhance the central release of GnRH and potentiate oestrogen-activated GnRH release. This plasticity involves dynamic cell interactions that bring into play tanycytes, astrocytes, vascular endothelial cells and GnRH neurones themselves. The underlying signalling pathways responsible for these structural changes are comprised of highly diffusible gaseous molecules, such as endothelial nitric oxide, and paracrine communication processes involving receptors of the erbB tyrosine kinase family, transforming growth factor beta 1 and eicosanoids, such as prostaglandin E(2). Some of these molecules, as a result of their ability to diffuse within the median eminence, may also serve as synchronizing cues allowing for the occurrence of functionally meaningful episodes of GnRH secretion by coordinating GnRH release from the GnRH neuroendocrine terminals.


Subject(s)
Endothelial Cells/metabolism , Gonadotropin-Releasing Hormone/metabolism , Median Eminence/metabolism , Nerve Endings/metabolism , Neurons/metabolism , Neurosecretory Systems/metabolism , Reproduction/physiology , Dinoprostone/metabolism , Endothelial Cells/cytology , Median Eminence/cytology , Neuroglia/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism
9.
Zoonoses Public Health ; 54(8): 320-7, 2007.
Article in English | MEDLINE | ID: mdl-17894643

ABSTRACT

Botulism is a rare but serious paralytic illness caused by a nerve toxin that is produced by the bacterium Clostridium botulinum. The economic, medical and alimentary consequences can be catastrophic in case of an epizooty. A polymerase chain reaction (PCR)-based assay was developed for the detection of C. botulinum toxigenic strains type C and D in bovine samples. This assay has proved to be less expensive, faster and simpler to use than the mouse bioassay, the current reference method for diagnosis of C. botulinum toxigenic strains. Three pairs of primers were designed, one for global detection of C. botulinum types C and D (primer pair Y), and two strain-specific pairs specifically designed for types C (primer pair VC) and D (primer pair VD). The PCR amplification conditions were optimized and evaluated on 13 bovine and two duck samples that had been previously tested by the mouse bioassay. In order to assess the impact of sample treatment, both DNA extracted from crude samples and three different enrichment broths (TYG, CMM, CMM followed by TYG) were tested. A 100% sensitivity was observed when samples were enriched for 5 days in CMM followed by 1 day in TYG broth. False-negative results were encountered when C. botulinum was screened for in crude samples. These findings indicate that the current PCR is a reliable method for the detection of C. botulinum toxigenic strains type C and D in bovine samples but only after proper enrichment in CMM and TYG broth.


Subject(s)
Botulism/veterinary , Cattle Diseases/diagnosis , Clostridium botulinum type C/isolation & purification , Clostridium botulinum type D/isolation & purification , Animals , Botulism/diagnosis , Cattle , Cattle Diseases/microbiology , Clostridium botulinum type C/genetics , Clostridium botulinum type D/genetics , DNA Primers , DNA, Bacterial/analysis , Mice , Polymerase Chain Reaction/veterinary , Predictive Value of Tests , Sensitivity and Specificity
10.
J Neuroendocrinol ; 19(9): 691-702, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17680884

ABSTRACT

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.


Subject(s)
Astrocytes , Gonadotropin-Releasing Hormone/analysis , Hypothalamus , Neurons , Adult , Aged , Aged, 80 and over , Animals , Astrocytes/chemistry , Astrocytes/cytology , Cell Shape , Female , Glial Fibrillary Acidic Protein/analysis , Humans , Hypothalamus/anatomy & histology , Hypothalamus/chemistry , Intermediate Filament Proteins/analysis , Nerve Tissue Proteins/analysis , Nestin , Neuronal Plasticity , Neurons/chemistry , Neurons/cytology , Vimentin/analysis
11.
Oncogene ; 26(19): 2695-706, 2007 Apr 26.
Article in English | MEDLINE | ID: mdl-17057735

ABSTRACT

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.


Subject(s)
Astrocytes/cytology , Cell Differentiation/drug effects , Neurons/metabolism , Stem Cells/cytology , Transforming Growth Factor alpha/pharmacology , Animals , Astrocytes/metabolism , Cell Lineage , Cell Movement , Cells, Cultured , Electrophysiology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , ErbB Receptors/metabolism , Female , Fetus/cytology , Fetus/metabolism , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL , Neuroglia/cytology , Neurons/cytology , Receptor, ErbB-2/metabolism , Recombinant Proteins/pharmacology , Stem Cells/metabolism
12.
Oncogene ; 25(29): 4076-85, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16532035

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Down-Regulation/drug effects , ErbB Receptors/metabolism , MAP Kinase Signaling System/drug effects , Receptor, ErbB-2/metabolism , Transforming Growth Factor alpha/pharmacology , Aging/metabolism , Animals , Astrocytes/cytology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/embryology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/metabolism , Humans , Mice , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Time Factors , Transforming Growth Factor alpha/metabolism
13.
Neurol Res ; 25(6): 635-41, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14503018

ABSTRACT

Chronic brain hypoperfusion (CBH) using permanent occlusion of both common carotid arteries in an aging rat model, has been shown to mimic human mild cognitive impairment (MCI), an acknowledged high risk condition that often converts to Alzheimer's disease. An aging rat model was used to determine whether hippocampal nitric oxide (NO) is abnormally expressed following CBH for two or eight weeks. At each time point, spatial memory was measured with the Morris water maze and hippocampal A beta 1-40/1-42 concentrations were obtained using sandwich ELISA. Real-time amperometric measures of NO representing the constitutive isoforms of neuronal nitric oxide synthase (nNOS) and endothelial (e)NOS were also taken at each time point to ascertain whether NO levels changed as a result of CBH, and if so, whether such NO changes preceded or followed any memory or amyloid-beta pathology. We found that two weeks after CBH, NO hippocampal levels were upregulated nearly four-fold when compared to nonoccluded rats but no alteration in spatial memory of A beta products were observed at this time point. By contrast, NO concentration had declined to control levels by eight weeks but spatial memory was found significantly impaired and A beta 1-40 (but not A beta 1-42) had increased in the CBH group when compared to control rats. Since changes in shear stress are known to upregulate eNOS but generally not nNOS, these results suggest that shear stress induced by CBH hyperactivated vascular NO derived from eNOS in the first two weeks as a reaction by the capillary endothelium to maintain homeostasis of local cerebral blood flow. The return of vascular NO to basal levels after eight weeks of CBH may have triggered metabolic changes within hippocampal cells resulting in hippocampal dysfunction as reflected by spatial memory impairment and by accumulation of A beta 1-40 peptide. In conclusion, our study shows that CBH initiates spatial memory loss in aging rats thus mimicking human MCI and also increases A beta 1-40 in the hippocampus. The memory and amyloid changes are preceded by NO upregulation in the hippocampus. These preliminary findings may be important in understanding, at least in part, the molecular mechanisms that precede memory impairment during chronic brain ischemia and as such, the pre-clinical stage leading to Alzheimer's disease.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Cerebrovascular Disorders/metabolism , Hippocampus/metabolism , Hypotension/metabolism , Memory Disorders/etiology , Nitric Oxide/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/physiopathology , Animals , Cerebrovascular Circulation/physiology , Cerebrovascular Disorders/complications , Cerebrovascular Disorders/physiopathology , Chronic Disease , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Hypotension/complications , Hypotension/physiopathology , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Up-Regulation/physiology
14.
Neuroscience ; 112(2): 475-85, 2002.
Article in English | MEDLINE | ID: mdl-12044465

ABSTRACT

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.


Subject(s)
Galanin/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Pro-Opiomelanocortin/metabolism , Animals , Calcineurin Inhibitors , Dose-Response Relationship, Drug , Galanin/antagonists & inhibitors , Hypothalamus/chemistry , Hypothalamus/cytology , Hypothalamus/physiology , In Situ Hybridization , In Vitro Techniques , Male , Neurons/chemistry , Neurons/cytology , Neurons/physiology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Receptors, Galanin , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tacrolimus/pharmacology , beta-Endorphin/analysis , beta-Endorphin/metabolism
15.
Endocrinology ; 142(10): 4288-94, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564686

ABSTRACT

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.


Subject(s)
Estrus/physiology , Gonadotropin-Releasing Hormone/physiology , Median Eminence/physiology , Nitric Oxide Synthase/physiology , Animals , Blood Vessels/physiology , Endothelium, Vascular/physiology , Female , Median Eminence/blood supply , Nitric Oxide Synthase Type III , Rats , Rats, Wistar
16.
Endocrinology ; 142(9): 4055-65, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11517185

ABSTRACT

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.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Drug Synergism , Estradiol/pharmacology , Female , Hormones/blood , Ovariectomy , Progesterone/pharmacology , Rats , Rats, Wistar , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology
17.
Endocrinology ; 142(6): 2343-50, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356681

ABSTRACT

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.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Median Eminence/metabolism , Nitric Oxide/metabolism , Animals , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrus , Female , In Vitro Techniques , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Ornithine/analogs & derivatives , Ornithine/pharmacology , Ovariectomy , Periodicity , Rats , Rats, Wistar
18.
Int J Mol Med ; 7(2): 119-29, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11172614

ABSTRACT

It is widely assumed that all exercise, regardless of the degree of difficulty or strenuousness, is good (no pain-no gain). In this speculative review of the literature and our research findings we highlight the fact that strenuous exercise taken to the extreme initiates an immune and vascular proinflammatory situation. However, mild cyclic exercise appears to produce health benefits for an individual. In part, this is due to vascular cyclic pulsations, occurring in mild exercise, stimulating constitutive nitric oxide synthase derived nitric oxide release. This in turn down-regulates vascular endothelial cells and immunocytes, as well as their interaction and inhibits the disassociation of NF-kappaB, preventing the production of proinflammatory cytokines. The nitric oxide so generated may even scavenge excess free radicals, preventing tissue damage. Prolonged strenuous exercise appears to limit these positive phenomena because of the maintained and prolonged high blood pressure that reduces the cyclic pulsations, limiting nitric oxide production. We further note that pathological conditions, i.e., Parkinson's disorder, may benefit from mild exercise, i.e., cyclic nitric oxide production, since the inactivity associated with this disease may lead to compromised nitric oxide production, initiating a progressive deterioration of tissues, including peripheral adrenergic neurons, due to a lack of adequate basal nitric oxide levels required to maintain the vascular microenvironment in a mild state of inhibition. We conclude that mild exercise represents an alternate and economical therapy to preserve health and/or diminish the rate of decline of the normal physiological processes that may even be associated with aging.


Subject(s)
Exercise/physiology , Nitric Oxide/blood , Pulsatile Flow/physiology , Humans , Periodicity , Vascular Diseases/etiology , Vascular Diseases/prevention & control
19.
Brain Res Brain Res Rev ; 34(1-2): 27-41, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11086185

ABSTRACT

It is becoming increasingly clear that nitric oxide (NO), an active free radical formed during the conversion of arginine to citrulline by the enzyme NO synthase (NOS), is a critical neurotransmitter and biological mediator of the neuroendocrine axis. Current evidence suggests that NO modulates the activity of both the hypothalamic-pituitary-gonadal axis and the hypothalamic-pituitary-adrenal axis. Supporting this hypothesis is the finding that the highest expression of neuronal NOS in the brain is found within the hypothalamus in areas where the cell bodies of the neurons from the different neuroendocrine systems are located. In this regard, the influence of neuronal NO on the regulation of the neuroendocrine neural cell body activity has been well-documented whereas little is known about NO signaling that directly modulates neurohormonal release into the pituitary portal vessels from the neuroendocrine terminals within the median eminence, the common termination field of the adenohypophysiotropic systems. Studies in rat suggest that NO is an important factor controlling both gonadotropin-releasing hormone (GnRH) and corticotropin-releasing hormone (CRH) release at the median eminence. The recent use of amperometric NO detection from median eminence fragments coupled to the use of selective NOS inhibitors demonstrated that a major source of NO at the median eminence might be endothelial in origin rather than neuronal. The present article reviews the recent progress in identifying the origin and the role of the NO produced at the median eminence in the control of neurohormonal release. We also discuss the potential implications of the putative involvement of the median eminence endothelial cells in a neurovascular regulatory process for hypothalamic neurohormonal signaling.


Subject(s)
Median Eminence/physiology , Nitric Oxide/physiology , Signal Transduction , Animals , Corticotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Neurosecretory Systems/physiology
20.
Recent Prog Horm Res ; 55: 197-223; discussion 223-4, 2000.
Article in English | MEDLINE | ID: mdl-11036938

ABSTRACT

It is becoming increasingly clear that astroglial cells are active participants in the process by which information is generated and disseminated within the central nervous system (CNS). In the hypothalamus, astrocytes regulate the secretory activity of neuroendocrine neurons. They contribute to facilitating sexual development by stimulating the release of luteinizing hormone-releasing hormone (LHRH), the neuropeptide that controls sexual development, from LHRH neurons. Astrocytes secrete several growth factors able to stimulate LHRH secretion. Two members of the epidermal growth factor (EGF) family--transforming growth factor alpha (TGFalpha) and the neuregulins (NRGs)-are produced in hypothalamic astrocytes and elicit LHRH secretion indirectly, via activation of receptor complexes formed by three members of the EGF receptor family, also located on astrocytes. Activation of these receptors results in the production of at least one neuroactive substance, prostaglandin E2 (PGE2), which stimulates LHRH secretion upon binding to specific receptors on LHRH neurons. Overexpression of TGFalpha in the hypothalamus accelerates puberty, whereas blockade of either TGFalpha or NRG actions delays the process, indicating that both peptides are physiological components of the neuroendocrine mechanism that controls sexual maturation. An increase in hypothalamic expression of at least two of the erbB receptors is initiated before the pubertal augmentation of gonadal steroid secretion and is completed on the day of the first preovulatory surge of gonadotropins. This secondary increase is brought about by gonadal steroids. Estrogen and progesterone facilitate erbB-mediated glia-to-LHRH neuron communication by enhancing astrocytic gene expression of at least one of the EGF-related ligands (TGFalpha) and two of the receptors (erbB-2 and erbB-4). They also facilitate the LHRH response to PGE2 via induction of PGE2 receptors in LHRH neurons. A search for genes that may act as upstream regulators of the pubertal process resulted in the identification of two potential candidates: Oct-2, a POU domain gene originally described in cells of the immune system, and TTF-1, a member of the Nkx family of homeodomain transcriptional regulators required for diencephalic morphogenesis. The hypothalamic expression of both genes increases during juvenile development before the first hormonal manifestations of puberty take place. Their mRNA transcripts are localized to specific hypothalamic cellular subsets, where they appear to regulate different, but interactive, components of the neuronal-glial complex controlling LHRH secretion. While Oct-2 transactivates the TGFalpha promoter, TTF-1 does so to the erbB-2 and LHRH genes but inhibits preproenkephalin promoter activity, suggesting that both transcriptional regulators may act coordinately in the normal hypothalamus to activate genes involved in facilitating the advent of puberty and repress those restraining sexual development. Altogether, these observations indicate that the central activation of the pubertal process involves the participation of both neuronal and astroglial networks and the contribution of upstream transcriptional regulators acting on both the neuronal and glial components of the system.


Subject(s)
Neurosecretory Systems/physiology , Sexual Maturation/physiology , Animals , Astrocytes/physiology , ErbB Receptors/physiology , Female , Gonadotropin-Releasing Hormone/metabolism , Models, Neurological , Neuroglia/physiology , Neurons/physiology , Ovary/physiology , Sexual Maturation/genetics , Signal Transduction , Steroids/physiology , Transforming Growth Factor alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...