Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell Death Differ ; 15(9): 1450-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18437159

ABSTRACT

Mouse embryonic stem (ES) cells remain pluripotent in vitro when grown in the presence of leukemia inhibitory factor (LIF) cytokine. LIF starvation leads to cell commitment, and part of the ES-derived differentiated cells die by apoptosis together with caspase3-cleavage and p38alpha activation. Inhibition of p38 activity by chemical compounds (PD169316 and SB203580), along with LIF withdrawal, leads to different outcomes on cell apoptosis, giving the opportunity to study the influence of apoptosis on cell differentiation. By gene profiling studies on ES-derived differentiated cells treated or not with these inhibitors, we have characterized the common and specific set of genes modulated by each inhibitor. We have also identified key genes that might account for their different survival effects. In addition, we have demonstrated that some genes, similarly regulated by both inhibitors (upregulated as Bcl2, Id2, Cd24a or downregulated as Nodal), are bona fide p38alpha targets involved in neurogenesis and found a correlation with their expression profiles and the onset of neuronal differentiation triggered upon retinoic acid treatment. We also showed, in an embryoid body differentiation protocol, that overexpression of EGFP (enhanced green fluorescent protein)-BCL2 fusion protein and repression of p38alpha are essential to increase formation of TUJ1-positive neuronal cell networks along with an increase in Map2-expressing cells.


Subject(s)
Embryonic Stem Cells/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Apoptosis , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Gene Expression/drug effects , Imidazoles/pharmacology , Mice , Neurons/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridines/pharmacology , Transcription, Genetic , Tretinoin/pharmacology
2.
Prog Neurobiol ; 60(4): 363-84, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10670705

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia in the elderly population. Dementia is associated with massive accumulation of fibrillary aggregates in various cortical and subcortical regions of the brain. These aggregates appear intracellularly as neurofibrillary tangles, extracellularly as amyloid plaques and perivascular amyloid in cerebral blood vessels. The causative factors in AD etiology implicate both, genetic and environmental factors. The large majority of early-onset familial Alzheimer's disease (FAD) cases are linked to mutations in the genes coding for presenilin 1 (PS1) and presenilin 2 (PS2). The corresponding proteins are 467 (PS1) and 448 (PS2) amino-acids long, respectively. Both are membrane proteins with multiple transmembrane regions. Presenilins show a high degree of conservation between species and a presenilin homologue with definite conservation of the hydrophobic structure has been identified even in the plant Arabidopsis thaliana. More than 50 missense mutations in PS1 and two missense mutations in PS2 were identified which are causative for FAD. PS mutations lead to the same functional consequence as mutations on amyloid precursor protein (APP), altering the processing of APP towards the release of the more amyloidogenic form 1-42 of Abeta (Abeta42). In this regard, the physical interaction between APP and presenilins in the endoplasmic reticulum has been demonstrated and might play a key role in Abeta42 production. It was hypothesized that PS1 might directly cleave APP. However, extracellular amyloidogenesis and Abeta production might not be the sole factor involved in AD pathology and several lines of evidence support a role of apoptosis in the massive neuronal loss observed. Presenilins were shown to modify the apoptotic response in several cellular systems including primary neuronal cultures. Some evidence is accumulating which points towards the beta-catenin signaling pathways to be causally involved in presenilin mediated cell death. Increased degradation of beta-catenin has been shown in brain of AD patients with PS1 mutations and reduced beta-catenin signaling increased neuronal vulnerability to apoptosis in cell culture models. The study of presenilin physiological functions and the pathological mechanisms underlying their role in pathogenesis clearly advanced our understanding of cellular mechanisms underlying the neuronal cell death and will contribute to the identification of novel drug targets for the treatment of AD.


Subject(s)
Alzheimer Disease/physiopathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Animals , Female , Humans , Pregnancy , Presenilin-1 , Presenilin-2
3.
Cell Death Dis ; 6: e1594, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25590804

ABSTRACT

Brain thiamine homeostasis has an important role in energy metabolism and displays reduced activity in Alzheimer's disease (AD). Thiamine deficiency (TD) induces regionally specific neuronal death in the animal and human brains associated with a mild chronic impairment of oxidative metabolism. These features make the TD model amenable to investigate the cellular mechanisms of neurodegeneration. Once activated by various cellular stresses, including oxidative stress, PKR acts as a pro-apoptotic kinase and negatively controls the protein translation leading to an increase of BACE1 translation. In this study, we used a mouse TD model to assess the involvement of PKR in neuronal death and the molecular mechanisms of AD. Our results showed that the TD model activates the PKR-eIF2α pathway, increases the BACE1 expression levels of Aß in specific thalamus nuclei and induces motor deficits and neurodegeneration. These effects are reversed by PKR downregulation (using a specific inhibitor or in PKR knockout mice).


Subject(s)
Amyloid beta-Peptides/biosynthesis , Down-Regulation , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Thiamine/metabolism , eIF-2 Kinase/metabolism , Amyloid/metabolism , Animals , Brain/enzymology , Brain/pathology , Caspase 3/metabolism , Disease Models, Animal , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Humans , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Motor Activity , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Protein Transport , Signal Transduction , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/deficiency
4.
Hum Gene Ther ; 10(18): 2987-97, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10609659

ABSTRACT

Huntington's disease (HD) is a genetic disorder leading to the degeneration of striatal GABA-ergic output neurons. No treatment is currently available for this devastating disorder, although several neurotrophic factors, including brain-derived neurotrophic factor (BDNF), have been shown to be beneficial for striatal neuron survival. We analyzed the effect of adenovirus-mediated transfer of the BDNF gene in a model of HD. Using a stereological procedure, three groups of rats were given an intrastriatal injection of adenovirus encoding BDNF, beta-galactosidase, or sham surgery. Two weeks after treatment, the animals were lesioned with quinolinic acid (QUIN), a toxin that induces striatal neuron death by an excitotoxic process. One month after the lesion, histological study revealed that striatal neurons were protected only in rats treated with the BDNF adenovirus. Volume measurements showed that the QUIN-induced lesions were 55% smaller in the BDNF adenovirus-treated group than in the beta-galactosidase adenovirus-treated group (p < 0.05), and the sham-treated group (p < 0.05). To determine the survival of striatal GABA-ergic output neurons after the QUIN-induced lesion, we immunostained brain sections with DARPP-32, an antibody specific for striatal output neurons. Prior treatment with the BDNF adenovirus resulted in a cell survival of 64%, whereas that after beta-galactosidase treatment was 46% (p < 0.05), showing that the BDNF adenovirus protected the striatal neurons. These results indicate that transfer of the BDNF gene is of therapeutic value for Huntington's disease.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Corpus Striatum/pathology , Gene Transfer Techniques , Huntington Disease/therapy , Adenoviridae/genetics , Animals , Base Sequence , Brain-Derived Neurotrophic Factor/genetics , DNA Primers , Disease Models, Animal , Female , Genetic Therapy , Genetic Vectors , Huntington Disease/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics
5.
FEBS Lett ; 383(3): 219-22, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8925899

ABSTRACT

At least 22 different mutations associated with early-onset familial Alzheimer's disease (AD) in various kindreds have been reported to occur in a recently identified gene on chromosome 14, presenilin 1 (PS-1) (Sherrington et al. (1995) Nature 375, 754-760 [1] and reviewed by Van Broeckhoven (1995) Nat. Genet. 11, 230-231 [2]). In order to study the localization of PS-1 in the brain, we raised a polyclonal antiserum specific to a fragment of the predicted protein sequence of PS-1. PS-1 immunostaining was found intracellularly, in the perikaria of discrete cells, mostly neurons, appearing as thick granules, resembling large-size vesicles. These granules were located in the periphery of cell bodies and extended into dendrites and neurites. PS-1 expression was found to be broadly distributed throughout the mouse brain, not only in structures involved in AD pathology, but also in structures unaltered by this disease.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Membrane Proteins/biosynthesis , Alzheimer Disease/genetics , Amino Acid Sequence , Animals , Brain/cytology , Chromosomes, Human, Pair 14 , Gene Expression , Humans , Immunohistochemistry , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Presenilin-1 , Protein Biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Transcription, Genetic
6.
J Comp Neurol ; 432(2): 184-96, 2001 Apr 02.
Article in English | MEDLINE | ID: mdl-11241385

ABSTRACT

The etiology of Parkinson's disease is unknown, but the gene involved in an autosomic recessive form of the disease with early onset has recently been identified. It codes for a protein with an unknown function called parkin. In the present study we produced a specific polyclonal antiserum against human parkin. Immunohistochemical analysis showed that parkin is expressed in neuronal perikarya and processes but also in glial and blood vessels in the primate brain (human and monkey). Electron microscopy indicated that parkin immunoreactivity is mostly located in large cytoplasmic vesicles and at the level of the endoplasmic reticulum. Parkin was expressed heterogeneously in various structures of the brain. It was detectable in the dopaminergic systems at the level of the perikarya in the mesencephalon but also in the striatum. However, parkin was also expressed by numerous nondopaminergic neurons. The staining intensity of parkin was particularly high in the hippocampal formation, the pallidal complex, the red nucleus, and the cerebellum. Comparison of control subjects with patients with Parkinson's disease and control animals with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated animals revealed a loss of parkin-immunoreactive neurons only in the substantia nigra pars compacta. Furthermore, the surviving dopaminergic neurons in the parkinsonian state continued to express parkin at a level similar to that observed in the control situation. These data indicate that parkin is a widely expressed protein. Thus, the degeneration of dopaminergic neurons in familial cases of Parkinson's disease with autosomal recessive transmission cannot be explained solely in terms of an alteration of this protein.


Subject(s)
Brain/metabolism , Ligases/metabolism , Neuroglia/metabolism , Neurons/metabolism , Parkinsonian Disorders/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adult , Aged , Aged, 80 and over , Animals , Antibodies/metabolism , COS Cells , Callithrix , Chlorocebus aethiops , Dopamine Agents , Endothelium, Vascular/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Parkinsonian Disorders/chemically induced , Substantia Nigra/metabolism , Ubiquitin-Protein Ligases
7.
Biochem Soc Symp ; (67): 81-8, 2001.
Article in English | MEDLINE | ID: mdl-11447842

ABSTRACT

Neurofibrillary tangles (NFTs) are a characteristic neuropathological lesion of Alzheimer's disease (AD). They are composed of a highly-phosphorylated form of the microtubule-associated protein tau. We are investigating the relationship between NFTs and microtubule stability and how tau phosphorylation and function is affected in transgenic models and by co-expression with beta-amyloid precursor protein and presenilins. In most NFT-bearing neurons, we observed a strong reduction in acetylated alpha-tubulin immunoreactivity (a marker of stable microtubules) and a reduction of the in situ hybridization signal for tubulin mRNA. In transfected cells, mutated tau forms (corresponding to tau mutations identified in familial forms of frontotemporal dementias linked to chromosome 17) were less efficient in their ability to sustain microtubule growth. These observations are consistent with the hypothesis that destabilization of the microtubule network is an important mechanism of cell dysfunction in Alzheimer's disease. The glycogen synthase kinase-3 beta (GSK-3 beta) generates many phosphorylated sites on tau. We performed a neuroanatomical study of GSK-3 beta distribution showing that developmental evolution of GSK-3 beta compartmentalization in neurons paralleled that of phosphorylated tau. Studies on transfected cells and on cultured neurons showed that GSK-3 beta activity controls tau phosphorylation and tau functional interaction with microtubules. Tau phosphorylation was not affected in neurons overexpressing beta-amyloid precursor protein. Transgenic mice expressing a human tau isoform and double transgenic animals for tau and mutated presenilin 1 have been generated; a somatodendritic accumulation of phosphorylated transgenic tau proteins, as observed in the pretangle stage in AD, has been observed but NFTs were not found, suggesting that additional factors might be necessary to induce their formation.


Subject(s)
Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , CHO Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cricetinae , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microtubules/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Presenilin-1 , Transfection , tau Proteins/genetics
8.
Neuropharmacology ; 35(5): 523-30, 1996 May.
Article in English | MEDLINE | ID: mdl-8887960

ABSTRACT

Glutamic acid is the major excitatory amino acid of the central nervous system which interacts with two receptor families, the ionotropic and metabotropic glutamate receptors. The metabotropic glutamate receptors (mGluRs) are coupled to G proteins and can be divided into three subgroups based on their sequence homology, signal transduction pathway and pharmacology. In this study, we describe the cloning of the cDNA encoding the human metabotropic glutamate receptor type 3 (HmGluR3). It was obtained by reverse transcription-polymerase chain reaction (RT-PCR) with degenerate oligonucleotides corresponding to highly conserved sequences between rat mGluRs. The receptor shows 879 amino acids with 96% amino acid sequence identity with rat mGluR3. It is strongly expressed in fetal and adult whole brain, especially in caudate nucleus and corpus callosum. The gene was identified by fluorescence in situ hybridization on chromosome 7 band q22. Activation of the human mGluR3, permanently expressed in Baby Hamster Kidney (BHK) cells, by excitatory amino acid inhibits the forskolin-stimulated accumulation of intracellular cAMP. The rank order of potency is L-glutamic acid > or = (1S,3R)-1-aminocyclopentane-1,3-dicarboxylic acid (1S,3R)-ACPD) >> ibotenic acid > quisqualic acid. (RS)-alpha-methyl-4-carboxyphenylglycine [(RS)-MCPG, 1 mM] is without effect on inhibition of forskolin-induced cAMP accumulation by L-glutamic acid.


Subject(s)
Cyclic AMP/metabolism , Glutamic Acid/pharmacology , Receptors, Metabotropic Glutamate/genetics , Amino Acid Sequence , Animals , Cricetinae , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Rats
9.
Neuroscience ; 95(2): 353-8, 2000.
Article in English | MEDLINE | ID: mdl-10658614

ABSTRACT

We investigated the effect of the noradrenergic neurotoxin, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) (1 or 3 x 50 mg/kg, intraperitoneally), on hippocampal, cortical and cerebellar noradrenaline levels after recovery of one, five and 11 months in control and apolipoprotein E-deficient mice. Apolipoprotein E-deficient mice had lower hippocampal noradrenaline levels than control mice. DSP-4-lesioned control mice had a more extensive recovery of hippocampal and cortical noradrenaline levels than DSP-4-lesioned apoE-deficient mice after five months' survival. Furthermore, the hippocampal noradrenaline levels after five and 11 months and cortical noradrenaline levels after five months of recovery had slightly recovered in control but not in apolipoprotein E-deficient mice treated with a single dose of DSP-4 compared with mice treated with three doses of DSP-4. These results show that apolipoprotein E-deficient mice have impaired recovery capacity in their locus coeruleus neurons.


Subject(s)
Apolipoproteins E/genetics , Nerve Degeneration/metabolism , Norepinephrine/blood , Alzheimer Disease/metabolism , Analysis of Variance , Animals , Benzylamines , Cerebellum/metabolism , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/enzymology , Frontal Lobe/metabolism , Hippocampus/metabolism , Mice , Mice, Mutant Strains , Nerve Degeneration/chemically induced , Serotonin/metabolism , Sympathomimetics
10.
Neuroscience ; 96(2): 291-7, 2000.
Article in English | MEDLINE | ID: mdl-10683569

ABSTRACT

We investigated whether the nucleus basalis lesion induced by quisqualic acid was associated with a more severe impairment of spatial navigation in a water maze, a greater reduction in frontal choline acetyltransferase activity and decrease in the number of choline acetyltransferase-positive neurons in the nucleus basalis in apolipoprotein E-deficient mice than in control mice. We also studied the effect of ageing on water maze spatial navigation and cortical choline acetyltransferase activity in 16-month-old control and apolipoprotein E-deficient mice. We found that the lesion decreased choline acetyltransferase-positive neurons in the nucleus basalis and frontal choline acetyltransferase activity equally in control and apolipoprotein E-deficient mice. The nucleus basalis lesion had no effect on the initial acquisition in the water maze in control and apolipoprotein E-deficient mice after 25 or 106 days of recovery. However, the nucleus basalis lesion impaired the reversal learning in the water maze similarly in both strains after 25 days of recovery, but had no effect after 106 days of recovery. Finally, water maze spatial navigation and cortical choline acetyltransferase activity were similar in old control and apolipoprotein E-deficient mice. These results suggest that young and old apolipoprotein E-deficient mice do not have impairments in cholinergic activity or spatial navigation. Furthermore, apolipoprotein E deficiency does not increase the sensitivity to cholinergic and spatial navigation deficits induced by lesioning of the nucleus basalis with an excitatory amino acid and does not slow down the behavioral recovery.


Subject(s)
Acetylcholine/deficiency , Apolipoproteins E/deficiency , Basal Nucleus of Meynert/physiopathology , Memory Disorders/physiopathology , Animals , Basal Nucleus of Meynert/pathology , Choline O-Acetyltransferase/analysis , Choline O-Acetyltransferase/deficiency , Denervation , Maze Learning/physiology , Mice , Neurons/pathology
11.
Neuroscience ; 87(2): 325-36, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9740395

ABSTRACT

Mutations in the gene for presenilin 1 are causative for the majority of cases of early onset familial Alzheimer's disease. Yet, the physiological function of presenilin 1 and the pathological mechanisms of the mutations leading to Alzheimer's disease are still unknown. To analyse potential pathological effects of presenilin 1 over-expression, we have generated transgenic rats which express high levels of human presenilin 1 protein in the brain. The over-expression of presenilin 1 leads to saturation of its normal processing and to the appearance of full-length protein in the transgenic rat brain. The transgenic protein is expressed throughout the brain and is predominantly found in neuronal cells. Cultured primary cortical neurons derived from these transgenic rats are significantly more sensitive than non-transgenic controls to apoptosis induced by standard culture conditions and to apoptosis induced by trophic factor withdrawal. Furthermore, the observed apoptosis is directly correlated with the expression of the transgenic protein. The results further emphasize the role of presenilin 1 in apoptotic cell death in native neuronal cultures.


Subject(s)
Alzheimer Disease/metabolism , Apoptosis/physiology , Membrane Proteins/analysis , Neurons/physiology , Animals , Animals, Genetically Modified , Blotting, Northern , Blotting, Western , Cells, Cultured , Female , Humans , Immunohistochemistry , Presenilin-1 , Rats , Rats, Inbred F344
12.
Brain Res Mol Brain Res ; 47(1-2): 108-16, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9221907

ABSTRACT

The evidence that betaA4 is central to the pathology of Alzheimer's disease (AD) came from the identification of several missense mutations in the amyloid precursor protein (APP) gene co-segregating with familial AD (FAD). In an attempt to study the proteolytical processing of mutated human APP in vivo, we have created transgenic mice expressing the human APP695 isoform with four FAD-linked mutations. Expression of the transgene was controlled by the promoter of the HMG-CR gene. Human APP is expressed in the brain of transgenic mice as shown by Western blot and immunohistology. The proteolytic processing of human APP in the transgenic mice leads to the generation of C-terminal APP fragments as well as to the release of betaA4. Despite substantial amounts of betaA4 detected in the brain of the transgenic mice, neither signs of Alzheimer's disease-related pathology nor related behavioural deficits could be demonstrated.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Mutation/genetics , Promoter Regions, Genetic/genetics , Proteins/metabolism , Animals , Immunohistochemistry , Mice , Mice, Transgenic
13.
J Neuroendocrinol ; 10(2): 101-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9535056

ABSTRACT

The amyloid precursor protein (APP) and APP-like (APLP) material, as visualized with the Mab22C11 antibody, have previously been shown to be associated with radial glia in hypothalamus, which are known to promote neurite outgrowth. By Northern blot analysis, APP 695 mRNA levels increased steadily over hypothalamic development, APP 770 mRNA was transiently expressed at 12 days postnatally, and APLP mRNA was only weakly expressed in the hypothalamus. The developmental pattern of APP moeities in mouse hypothalamus and in fetal hypothalamic neurons in culture was compared with a presenilin 2 (PS2) related protein using an antibody developed against the N-terminal part of PS2. By Western blot analysis, APP and PS2-like immunoreactivity were visualized as a 100-130 and 52 kDa bands, respectively. An APP biphasic increase was observed during hypothalamic development in vivo. APP immunoreactivity was equally detected in neuronal and glial cultures, while PS2-like material was more concentrated in neurons. A correlation between APP/APP-like and PS2-like levels was observed during development in vivo. While APP was mostly associated with membrane fractions, a significant portion of PS2-like material was also recovered from cytosolic fractions in vitro. In contrast to native PS2 in COS-transfected cells, the PS2-like material did not aggregate after heating for 90 s at 90 degrees C. These results indicate a close association between APP and PS2-like material during hypothalamic development in vivo, and suggest that neuronal and glial cultures may provide appropriate models to test their interactions.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Hypothalamus/metabolism , Hypothalamus/physiology , Membrane Proteins/metabolism , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Hypothalamus/embryology , Mice , Presenilin-2 , RNA, Messenger/metabolism
14.
Psychopharmacology (Berl) ; 145(4): 386-92, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10460315

ABSTRACT

RATIONALE: Recent studies suggest that apoE-deficient mice may have impaired central cholinergic function and neuronal recovery capacity. OBJECTIVE: We investigated whether apoE-deficient mice are more susceptible to the biochemical and EEG defects induced by ageing or nucleus basalis (NB) lesion. METHODS: ApoE-deficient and control mice were used. The baseline EEG activity and EEG response to a muscarinic acetylcholine receptor antagonist, scopolamine (0.05 and 0.2 mg/kg) and a benzodiazepine receptor agonist, diazepam (0.5 and 2.0 mg/kg), were studied during ageing. In addition, the cortical and hippocampal ChAT activities were measured in aged mice. The baseline EEG activity and EEG response to scopolamine (0.05 and 0.2 mg/kg), and cortical ChAT activity, were studied after quisqualic acid-induced unilateral NB lesion. RESULTS: The baseline EEG fast wave activity (relative alpha and beta) was higher in apoE-deficient mice. Ageing decreased relative alpha activity similarly in both strains. The scopolamine induced EEG slowing was less prominent in apoE-deficient than in control mice, and the difference between the strains became slightly clearer during ageing. The NB lesion failed to produce more severe changes in cortical EEG and ChAT activity in apoE-deficient mice. Cortical and hippocampal ChAT activity was equal in young and aged apoE-deficient and control mice. The EEG response to diazepam in young and aged mice was similar in both strains. CONCLUSIONS: The regulation of cortical EEG activity of apoE-deficient mice was somewhat altered during ageing and the response to scopolamine treatment was blunted. However, the cholinergic cells of the NB of apoE-deficient mice were not more sensitive to lesion or to ageing, suggesting that apoE does not have to be present to preserve the viability of cholinergic neurons.


Subject(s)
Aging/physiology , Apolipoproteins E/deficiency , Electroencephalography/drug effects , Prosencephalon/physiology , Scopolamine/pharmacology , Animals , Choline O-Acetyltransferase/metabolism , Diazepam/pharmacology , Male , Mice , Mice, Inbred C57BL
15.
Neuroreport ; 7(15-17): 2587-91, 1996 Nov 04.
Article in English | MEDLINE | ID: mdl-8981428

ABSTRACT

We examined the regional and cellular distribution of presenilin-1 gene expression in the rat brain by in situ hybridization. Microscopic analysis demonstrated that presenilin-1 mRNA is predominantly expressed in areas such as the occipital cortex, the pyramidal layer of the hippocampus, thalamic nuclei and the cerebellar granular layer. The expression of presenilin-1 is mostly neuronal: only a weak hybridization signal was found in the corpus callosum and in the astrocytoma cell lines U373MG and U138MG.


Subject(s)
Brain/metabolism , Membrane Proteins/metabolism , Animals , In Situ Hybridization , Male , Presenilin-1 , Rats
16.
Neuroreport ; 7(12): 2021-5, 1996 Aug 12.
Article in English | MEDLINE | ID: mdl-8905717

ABSTRACT

Presenilin 2 (STM2) is a recently cloned gene involved in some forms of early onset Alzheimer's disease with autosomal dominant inheritance. Here we report the regional and cellular distribution of STM2 mRNA in the normal human central nervous system. Using in situ hybridization. STM2 gene expression was shown to be confined exclusively to neurones in the central nervous system. A high level of STM2 mRNA expression was observed in the cerebral cortex and the hippocampus, more particularly on pyramidal neurones of Ammon's horn and granular neurones of the dentate gyrus. STM2 mRNA was also detected in Purkinje cells and granular cells of the cerebellum, and in neurones of the striatum and the nucleus basalis of Meynert. Taken together, these results suggest that the expression of STM2 mRNA is not restricted to the neuronal populations that are known to degenerate in Alzheimer's disease.


Subject(s)
Brain/metabolism , Gene Expression/genetics , Membrane Proteins/genetics , Aged , Female , Hippocampus/metabolism , Humans , In Situ Hybridization , Male , Middle Aged , Presenilin-2
17.
Brain Res ; 758(1-2): 209-17, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9203550

ABSTRACT

Missense mutations of presenilin 1 (PS-1) and presenilin 2 (PS-2) genes cause the majority of early-onset familial forms of Alzheimer's disease (AD). We previously characterized the distribution of the PS-1 protein in the mouse brain by immunohistochemistry using an antibody directed against an epitope located in the large hydrophilic loop [Moussaoui, S., Czech, C., Pradier, L., Blanchard, V., Bonici, B., Gohin, M., Imperato, A. and Revah, F., Immunohistochemical analysis of presenilin 1 expression in the mouse brain, FEBS Lett., 383 (1996) 219-222]. Similarly, we now report the distribution pattern of PS-2 protein in the mouse brain. For these experiments we used a polyclonal antibody raised against a synthetic peptide corresponding to the amino-acid sequence 7-24 of the predicted human PS-2 protein. The specificity of the antibody was evidenced by its ability to recognize PS-2 protein in immunoprecipitation studies and by antigen-peptide competition. In the mouse brain, PS-2 protein was present in numerous cerebral structures, but its distribution in these structures did not correlate with their susceptibility to AD pathology. In all examined structures of the gray matter, PS-2 protein was concentrated in neuronal cell bodies but it was not detected in the glial cells of the white matter. The regional distribution pattern of PS-2 protein was almost identical to that of PS-1 protein. Moreover, PS-2 protein co-localized with PS-1 protein in a large number of neuronal cell bodies. In terms of subcellular localization, PS-2 immunostaining was present almost exclusively in neuronal cell bodies while PS-1 immunostaining was also present in dendrites. This could be explained by the different epitopes of the antibodies and the known proteolytic processing of both presenilins in vivo [Tanzi, R.E., Kovacs, D.M., Kim, T.-W., Moir, R.D., Guenette, S.Y. and Wasco, W., The presenilin genes and their role in early-onset familial Alzheimer's disease, Alzheimer's disease Rev., 1 (1996) 91-98]. Within neuronal cell bodies, the immunostaining of PS-2 protein, as well as that of PS-1 protein, had a reticular and granular appearance. This suggests in agreement with previous observations on PS-1 and PS-2 in COS and H4 cells [Kovacs, D.M., Fausett, H.J., Page, K.J., Kim, T.-W., Moir, R.D., Merriam, D.E., Hollister, R.D., Hallmark, O.G., Mancini, R., Felsenstein, K.M., Hyman, B.T., Tanzi, R.E., Wasco, W., Alzheimer-associated presenilins 1 and 2: neuronal expression in brain and localization to intracellular membranes in mammalian cells, Nature Med., 2 (1996) 224-229] that these proteins are situated in intracytoplasmic organelles, possibly the endoplasmic reticulum and the Golgi complex.


Subject(s)
Brain/metabolism , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Gene Expression/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Precipitin Tests , Presenilin-1 , Presenilin-2
18.
Eur J Pharmacol ; 235(2-3): 283-9, 1993 Apr 28.
Article in English | MEDLINE | ID: mdl-7685290

ABSTRACT

The effects of riluzole, an anticonvulsant and neuroprotective compound, on excitatory amino acid-evoked currents were studied in Xenopus laevis oocytes injected with mRNA from rat whole brain or cortex. Responses to kainic acid were blocked by riluzole (IC50 = 167 microM) as well as by the quinoxalinedione antagonists 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX: IC50 = 0.21 microM) and 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline (NBQX: IC50 = 0.043 microM). Riluzole was somewhat more potent at blocking responses to N-methyl-D-aspartic acid (NMDA: IC50 = 18.2 microM); the competitive NMDA receptor antagonist 2-amino-phosphonovaleric acid (2-APV) yielded an IC50 of 6.1 microM in this system. The inhibition by both riluzole and 2-APV was reversible and did not appear to be use dependent, unlike that of the channel blocker MK-801 ([+]-5-methyl-10,11-dihydro-5H-dibenzo-[a,d]cyclohepten-5,10-imine maleate). It was impossible to demonstrate an interaction of riluzole with any of the known ligand recognition sites on either the kainate or the NMDA receptor in radioligand binding studies. These results suggest a direct but non-competitive action of riluzole on ionotropic glutamate receptors.


Subject(s)
Ion Channels/drug effects , Kainic Acid/pharmacology , Oocytes/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Thiazoles/pharmacology , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Electrophysiology , Female , In Vitro Techniques , Kainic Acid/antagonists & inhibitors , RNA, Messenger/isolation & purification , Radioligand Assay , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Riluzole , Synaptic Transmission/drug effects , Xenopus laevis
19.
Eur J Pharmacol ; 245(1): 43-50, 1993 Mar 15.
Article in English | MEDLINE | ID: mdl-7682962

ABSTRACT

In the present study, we describe the effects of RP 67580, a substance P non-peptide antagonist, in binding and second messenger experiments performed using transfected Chinese hamster ovary cells expressing the rat NK1 receptor. The cDNA sequence encoding the rat brain substance P receptor was transfected in Chinese hamster ovary cells, and cellular clones which stably express the corresponding protein were isolated. [3H]Substance P binding was performed in homogenates of these transfected cells and revealed the presence of NK1 receptors in displacement experiments, using peptide analogs of three mammalian tachykinins (substance P, neurokinin A, neurokinin B). Scatchard analysis indicated a KD value of 0.33 +/- 0.13 nM and a Bmax value of 5.83 +/- 1.16 pmol/mg of protein. RP 67580, a selective NK1-receptor antagonist was found to displace the specific binding of [3H]substance P. When [3H]RP 67580 was used as a ligand, it displayed a high affinity (KD value: 1.22 +/- 0.27 nM) in transfected cell homogenates and only competed with NK1 receptor ligands. Substance P stimulated the hydrolysis of phosphoinositide in a time- and concentration-dependent manner and this effect was mimicked by selective agonists of the NK1 receptor ([Pro9]SP and septide). RP 67580 did not induce any accumulation of inositol phosphates, but was found to inhibit the inositol phosphate increase mediated by substance P, without affecting the maximal response. From these results, one may conclude that the receptor expressed by the transfected Chinese hamster ovary cells revealed similar binding characteristics as the NK1 receptor present in the rat brain and also confirmed the high affinity and the antagonist properties of RP 67580.


Subject(s)
Indoles/pharmacology , Receptors, Neurotransmitter/metabolism , Substance P/metabolism , Animals , CHO Cells , Cloning, Molecular , Cricetinae , Cricetulus , Hydrolysis , Inositol Phosphates/metabolism , Isoindoles , Rats , Receptors, Neurokinin-1 , Receptors, Neurokinin-2 , Receptors, Neurotransmitter/antagonists & inhibitors , Receptors, Neurotransmitter/genetics , Substance P/antagonists & inhibitors , Transfection
20.
Neurosci Lett ; 316(3): 145-8, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11744223

ABSTRACT

There is circumstantial evidence that the reelin signaling pathway may contribute to neurodegeneration in the adult brain and could be linked to Alzheimer's disease (AD). In the present immunohistochemical report we studied the reelin expression profile in double-transgenic mice that express both human mutant beta-amyloid precursor protein (APP) and human mutant presenilin-1. We were able to demonstrate that reelin immunostaining was found together with human APP in the neuritic component of many AD-typical plaques in both hippocampus and neocortex. This observation gives the first evidence for the association of reelin with amyloid deposits.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Plaque, Amyloid/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Extracellular Matrix Proteins/genetics , Female , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Interneurons/metabolism , Interneurons/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Presenilin-1 , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Reelin Protein , Serine Endopeptidases
SELECTION OF CITATIONS
SEARCH DETAIL