Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neural Plast ; 2022: 3172861, 2022.
Article in English | MEDLINE | ID: mdl-35237315

ABSTRACT

Recently, we showed that DNA double-strand breaks (DSBs) are increased by the Aß 42-amyloid peptide and decreased by all-trans retinoic acid (RA) in SH-SY5Y cells and C57BL/6J mice. The present work was aimed at investigating DSBs in cells and murine models of Alzheimer's disease carrying the preseniline-1 (PS1) P117L mutation. We observed that DSBs could hardly decrease following RA treatment in the mutated cells compared to the wild-type cells. The activation of the amyloidogenic pathway is proposed in the former case as Aß 42- and RA-dependent DSBs changes were reproduced by an α-secretase and a γ-secretase inhibitions, respectively. Unexpectedly, the PS1 P117L cells showed lower DSB levels than the controls. As the DSB repair proteins Tip60 and Fe65 were less expressed in the mutated cell nuclei, they do not appear to contribute to this difference. On the contrary, full-length BRCA1 and BARD1 proteins were significantly increased in the chromatin compartment of the mutated cells, suggesting that they decrease DSBs in the pathological situation. These Western blot data were corroborated by in situ proximity ligation assays: the numbers of BRCA1-BARD1, not of Fe65-Tip60 heterodimers, were increased only in the mutated cell nuclei. RA also enhanced the expression of BARD1 and of the 90 kDa BRCA1 isoform. The increased BRCA1 expression in the mutated cells can be related to the enhanced difficulty to inhibit this pathway by BRCA1 siRNA in these cells. Overall, our study suggests that at earlier stages of the disease, similarly to PS1 P117L cells, a compensatory mechanism exists that decreases DSB levels via an activation of the BRCA1/BARD1 pathway. This supports the importance of this pathway in neuroprotection against Alzheimer's disease.


Subject(s)
Alzheimer Disease , DNA Breaks, Double-Stranded , Alzheimer Disease/genetics , Animals , DNA , DNA Repair , Mice , Mice, Inbred C57BL , Presenilin-1/genetics
2.
Neural Plast ; 2020: 9369815, 2020.
Article in English | MEDLINE | ID: mdl-32256561

ABSTRACT

In this study, we have investigated the role of all-trans-retinoic acid (RA) as a neuroprotective agent against Aß 1-42-induced DNA double-strand breaks (DSBs) in neuronal SH-SY5Y and astrocytic DI TNC1 cell lines and in murine brain tissues, by single-cell gel electrophoresis. We showed that RA does not only repair Aß 1-42-induced DSBs, as already known, but also prevents their occurrence. This effect is independent of that of other antioxidants studied, such as vitamin C, and appears to be mediated, at least in part, by changes in expression, not of the RARα, but of the PPARß/δ and of antiamyloidogenic proteins, such as ADAM10, implying a decreased production of endogenous Aß. Whereas Aß 1-42 needs transcription and translation for DSB production, RA protects against Aß 1-42-induced DSBs at the posttranslational level through both the RARα/ß/γ and PPARß/δ receptors as demonstrated by using specific antagonists. Furthermore, it could be shown by a proximity ligation assay that the PPARß/δ-RXR interactions, not the RARα/ß/γ-RXR interactions, increased in the cells when a 10 min RA treatment was followed by a 20 min Aß 1-42 treatment. Thus, the PPARß/δ receptor, known for its antiapoptotic function, might for these short-time treatments play a role in neuroprotection via PPARß/δ-RXR heterodimerization and possibly expression of antiamyloidogenic genes. Overall, this study shows that RA can not only repair Aß 1-42-induced DSBs but also prevent them via the RARα/ß/γ and PPARß/δ receptors. It suggests that the RA-dependent pathways belong to an anti-DSB Adaptative Gene Expression (DSB-AGE) system that can be targeted by prevention strategies to preserve memory in Alzheimer's disease and aging.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Brain/metabolism , DNA Breaks, Double-Stranded , Neuroprotective Agents/administration & dosage , Peptide Fragments/toxicity , Tretinoin/administration & dosage , Tretinoin/metabolism , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , Humans , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects
3.
Neurobiol Dis ; 121: 95-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30261283

ABSTRACT

The involvement of the 18kDa translocator protein (TSPO), a marker of neuroinflammation, in Alzheimer's disease (AD) remains controversial. In the present report, we used [125I]-CLINDE, a SPECT TSPO radiotracer never before used in AD, and we investigated the relationship between TSPO and amyloid plaque density (using [125I]-DRM106) in a triple transgenic mouse model of AD (3xTgAD, APPSWE, PS1M146V and TauP301L). Our results show that TSPO increases appear before those of amyloid deposits. Moreover, the different parts of the hippocampus are differentially affected. Indeed, for both TSPO and amyloid, the subiculum is affected earlier and the ventral hippocampus later than the dorsal hippocampus. In the subiculum and the dorsal hippocampus of 3xTgAD mice, a positive correlation between TSPO and of amyloid deposit levels is observed. This data supports the hypothesis that TSPO could be used as a predictive marker of amyloid pathology. In addition, our immunohistochemical data shows a segregation of TSPO in the hippocampus and immunofluorescence imaging revealed a mainly microglial origin of the TSPO expression. Thus, imaging TSPO with CLINDE may be a good alternative to PET radiotracers.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Plaque, Amyloid/metabolism , Receptors, GABA/metabolism , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Animals , Choroid Plexus/metabolism , Disease Models, Animal , Encephalitis/complications , Encephalitis/metabolism , Female , Hippocampus/diagnostic imaging , Humans , Mice, Inbred C57BL , Mice, Transgenic , Tomography, Emission-Computed, Single-Photon/methods
5.
Neurodegener Dis ; 16(5-6): 398-406, 2016.
Article in English | MEDLINE | ID: mdl-27400224

ABSTRACT

BACKGROUND/AIMS: Investigations of Aß oligomers in neuropathologically confirmed Alzheimer's disease (AD) are still scarce. We report neurohistopathological and biochemical analyses using antibodies against tau and amyloid ß (Aß) pathology. METHODS: Thirty elderly AD patients and 43 age-matched controls with or without deposition of amyloid plaques (AP) were analyzed by immunohistochemistry. In 21 cases with available fresh tissue, Western blots were also performed. Neuropathological analysis included quantitative assessment of neurofibrillary tangles (NFT), AP and Aß oligomer densities in the mesial temporal cortex (TC). RESULTS: NFT, fibrillar amyloid and Aß oligomeric deposit densities were significantly higher in AD patients than in controls. There was no relationship between oligomeric Aß densities and Braak NFT staging scores. Furthermore, Aß oligomer expression was closely correlated with Aß plaques in the TC. By Western blot, Aß oligomers were observed in AD patients, in plaque-free controls, in 1 'tangle-only AD' case, as well as in the cerebellum. A band near 55 kDa was the only Western blot signal that was significantly increased in the TC of AD patients compared to controls as well as less expressed in the cerebellum. CONCLUSION: These results suggest that a putative dodecamer, near 55 kDa, may contribute to AD vulnerability of the TC.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cerebellum/metabolism , Temporal Lobe/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Cerebellum/pathology , Female , Humans , Male , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Temporal Lobe/pathology
6.
Neural Plast ; 2016: 3707406, 2016.
Article in English | MEDLINE | ID: mdl-26881107

ABSTRACT

The amyloid-ß peptide or Aß is the key player in the amyloid-cascade hypothesis of Alzheimer's disease. Aß appears to trigger cell death but also production of double-strand breaks (DSBs) in aging and Alzheimer's disease. All-trans retinoic acid (RA), a derivative of vitamin A, was already known for its neuroprotective effects against the amyloid cascade. It diminishes, for instance, the production of Aß peptides and their oligomerisation. In the present work we investigated the possible implication of RA receptor (RAR) in repair of Aß-induced DSBs. We demonstrated that RA, as well as RAR agonist Am80, but not AGN 193109 antagonist, repair Aß-induced DSBs in SH-SY5Y cells and an astrocytic cell line as well as in the murine cortical tissue of young and aged mice. The nonhomologous end joining pathway and the Ataxia Telangiectasia Mutated kinase were shown to be involved in RA-mediated DSBs repair in the SH-SY5Y cells. Our data suggest that RA, besides increasing cell viability in the cortex of young and even of aged mice, might also result in targeted DNA repair of genes important for cell or synaptic maintenance. This phenomenon would remain functional up to a point when Aß increase and RA decrease probably lead to a pathological state.


Subject(s)
Amyloid beta-Peptides/toxicity , DNA Breaks, Double-Stranded/drug effects , Neocortex/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Tretinoin/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/physiology , Neurons/physiology , Vitamin A/pharmacology
8.
Ageing Res Rev ; 18: 86-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25264360

ABSTRACT

The postsynaptic density protein PSD-95 is a major element of synapses. PSD-95 is involved in aging, Alzheimer's disease (AD) and numerous psychiatric disorders. However, contradictory data about PSD-95 expression in aging and AD have been reported. Indeed in AD versus control brains PSD-95 varies according to regions, increasing in the frontal cortex, at least in a primary stage, and decreasing in the temporal cortex. In contrast, in transgenic mouse models of aging and AD PSD-95 expression is decreased, in behaviorally aged impaired versus unimpaired rodents it can decrease or increase and finally, it is increased in rodents grown in enriched environments. Different factors explain these contradictory results in both animals and humans, among others concomitant psychiatric endophenotypes, such as depression. The possible involvement of PSD-95 in reactive and/or compensatory mechanisms during AD progression is underscored, at least before the occurrence of important synaptic elimination. Thus, in AD but not in AD transgenic mice, enhanced expression might precede the diminution commonly observed in advanced aging. A two-compartments cell model, separating events taking place in cell bodies and synapses, is presented. Overall these data suggest that AD research will progress by untangling pathological from protective events, a prerequisite for effective therapeutic strategies.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Age Factors , Aging/genetics , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Animals , Brain/pathology , Brain/physiopathology , Disks Large Homolog 4 Protein , Gene Expression Regulation , Genotype , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice, Transgenic , Models, Biological , Phenotype
9.
Neurobiol Aging ; 35(2): 322-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24011540

ABSTRACT

Superficial layers I to III of the human cerebral cortex are more vulnerable toward Aß peptides than deep layers V to VI in aging. Three models of layers were used to investigate this pattern of frailty. First, primary neurons from E14 and E17 embryonic murine cortices, corresponding respectively to future deep and superficial layers, were treated either with Aß(1-42), okadaic acid, or kainic acid. Second, whole E14 and E17 embryonic cortices, and third, in vitro separated deep and superficial layers of young and old C57BL/6J mice, were treated identically. We observed that E14 and E17 neurons in culture were prone to death after the Aß and particularly the kainic acid treatment. This was also the case for the superficial layers of the aged cortex, but not for the embryonic, the young cortex, and the deep layers of the aged cortex. Thus, the aged superficial layers appeared to be preferentially vulnerable against Aß and kainic acid. This pattern of vulnerability corresponds to enhanced accumulation of senile plaques in the superficial cortical layers with aging and Alzheimer's disease.


Subject(s)
Aging/pathology , Cell Survival/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurons/drug effects , Neurons/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/adverse effects , Animals , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Humans , Kainic Acid/adverse effects , Mice , Mice, Inbred C57BL , Okadaic Acid/adverse effects , Peptide Fragments/adverse effects , Plaque, Amyloid/metabolism
10.
Curr Alzheimer Res ; 11(1): 86-96, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24156266

ABSTRACT

In Alzheimer's disease (AD), synaptic alterations play a major role and are often correlated with cognitive changes. In order to better understand synaptic modifications, we compared alterations in NMDA receptors and postsynaptic protein PSD-95 expression in the entorhinal cortex (EC) and frontal cortex (FC; area 9) of AD and control brains. We combined immunohistochemical and image analysis methods to quantify on consecutive sections the distribution of PSD-95 and NMDA receptors GluN1, GluN2A and GluN2B in EC and FC from 25 AD and control cases. The density of stained receptors was analyzed using multivariate statistical methods to assess the effect of neurodegeneration. In both regions, the number of neuronal profiles immunostained for GluN1 receptors subunit and PSD-95 protein was significantly increased in AD compared to controls (3-6 fold), while the number of neuronal profiles stained for GluN2A and GluN2B receptors subunits was on the contrary decreased (3-4 fold). The increase in marked neuronal profiles was more prominent in a cortical band corresponding to layers 3 to 5 with large pyramidal cells. Neurons positive for GluN1 or PSD-95 staining were often found in the same localization on consecutive sections and they were also reactive for the anti-tau antibody AD2, indicating a neurodegenerative process. Differences in the density of immunoreactive puncta representing neuropile were not statistically significant. Altogether these data indicate that GluN1 and PSD-95 accumulate in the neuronal perikarya, but this is not the case for GluN2A and GluN2B, while the neuropile compartment is less subject to modifications. Thus, important variations in the pattern of distribution of the NMDA receptors subunits and PSD-95 represent a marker in AD and by impairing the neuronal network, contribute to functional deterioration.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Adult , Aged , Aged, 80 and over , Amyloid/metabolism , Disks Large Homolog 4 Protein , Entorhinal Cortex/metabolism , Female , Frontal Lobe/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/metabolism
11.
Brain Res Bull ; 94: 17-22, 2013 May.
Article in English | MEDLINE | ID: mdl-23396115

ABSTRACT

Patients with the early-onset Alzheimer's disease P117L mutation in the presenilin-1 gene (PS-1) present pathological hallmarks in the hippocampus, the frontal cortex and the basal ganglia. In the present work we determined by immunohistochemistry which brain regions were injured in the transgenic PS-1 P117L mice, in comparison to their littermates, the B6D2 mice. Furthermore, as these regions are involved in novelty detection, we investigated the behavior of these mice in tests for object and place novelty recognition. Limited numbers of senile plaques and neurofibrillary tangles were detected in aged PS-1 P117L mice in the CA1 only, indicating that the disease is restrained to an initial neuropathological stage. Western blots showed a change in PSD-95 expression (p=0.03), not in NR2A subunit, NR2B subunit and synaptophysin expressions in the frontal cortex, suggesting specific synaptic alterations. The behavioral tests repeatedly revealed, despite a non-significant preference for object or place novelty, maladaptive exploratory behavior of the PS-1 P117L mice in novel environmental conditions, not due to locomotor problems. These mice, unlike the B6D2 mice, were less inhibited to visit the center of the cages (p=0.01) and they continued to move excessively in the presence of a displaced object (p=0.021). Overall, the PS-1 P117L mice appear to be in an initial Alzheimer's disease-like neuropathological stage, and they showed a lack of reaction toward novel environmental conditions.


Subject(s)
Alzheimer Disease/genetics , Brain/pathology , Exploratory Behavior/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Blotting, Western , Brain/metabolism , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Transgenic , Mutation , Presenilin-1/genetics
12.
Exp Biol Med (Maywood) ; 237(11): 1331-40, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23239444

ABSTRACT

In the present work we studied synaptic protein concentrations in relation to behavioral performance. Long-Evans rats, aged 22-23 months, were classified for individual expression of place memory in the Morris water maze, in reference to young adults. Two main subgroups of aged rats were established: the Aged cognitively Unimpaired (AU) had search accuracy within the range (percent of time in training sector within mean ± 2 SEM) of young rats and the Aged cognitively Impaired (AI) rats had search accuracy below this range. Samples from the hippocampus and frontal cortex of all the AI, AU and young rats were analyzed for the expression of postsynaptic protein PSD-95 by Image J analysis of immunohistochemical data and by Western blots. PSD-95 expression was unchanged in the hippocampus, but, together with synaptophysin, was significantly increased in the frontal cortex of the AI rats. A significant correlation between individual accuracy (time spent in the training zone) and PSD-95 expression was observed in the aged group. No significant effect of age or PSD-95 expression was observed in the learning of a new position. All together, these data suggest that increased expression of PSD-95 in the frontal cortex of aged rats co-occurs with cognitive impairment that might be linked to functional alterations extending over frontal networks.


Subject(s)
Cognition Disorders/metabolism , Frontal Lobe/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Age Factors , Animals , Behavior, Animal/physiology , Cognition Disorders/pathology , Disks Large Homolog 4 Protein , Frontal Lobe/pathology , Hippocampus/metabolism , Maze Learning/physiology , Memory/physiology , Rats , Rats, Long-Evans , Synaptophysin/metabolism
13.
Brain Res Bull ; 80(4-5): 296-301, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19555743

ABSTRACT

The presenilin-1 gene is mutated in early-onset familial Alzheimer's disease. The mutation Pro117Leu is implicated in a very severe form of the disease, with an onset of less than 30 years. The consequences of this mutation on neurogenesis in the hippocampus of adult transgenic mice have already been studied in situ. The survival of neural progenitor cells was impaired resulting in decreased neurogenesis in the dentate gyrus. Our intention was to verify if similar alterations could occur in vitro in progenitor cells from the murine ganglionic eminences isolated from embryos of this same transgenic mouse model. These cells were grown in culture as neurospheres and after differentiation the percentage of neurons generated as well as their morphology were analysed. The mutation results in a significant decrease in neurogenesis compared to the wild type mice and the neurons grow longer and more ramified neurites. A shift of differentiation towards gliogenesis was observed that could explain decreased neurogenesis despite increased proliferation of neural precursors in transgenic neurospheres. A diminished survival of the newly generated mutant neurons is also proposed. Our data raise the possibility that these alterations in embryonic development might contribute to increase the severity of the Alzheimer's disease phenotype later in adulthood.


Subject(s)
Alzheimer Disease/genetics , Neurogenesis/genetics , Neurons/cytology , Presenilin-1/genetics , Animals , Apoptosis/genetics , Astrocytes/metabolism , Cell Count , Cells, Cultured , Immunohistochemistry , Mice , Mice, Transgenic , Multipotent Stem Cells/cytology , Mutation , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Brain Res Bull ; 80(4-5): 196-202, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19559767

ABSTRACT

In order to understand relationships between executive and structural deficits in the frontal cortex of patients within normal aging or Alzheimer's disease, we studied frontal pathological changes in young and old controls compared to cases with sporadic (AD) or familial Alzheimer's disease (FAD). We performed a semi-automatic computer assisted analysis of the distribution of beta-amyloid (Abeta) deposits revealed by Abeta immunostaining as well as of neurofibrillary tangles (NFT) revealed by Gallyas silver staining in Brodman areas 10 (frontal polar), 12 (ventro-infero-median) and 24 (anterior cingular), using tissue samples from 5 FAD, 6 sporadic AD and 10 control brains. We also performed densitometric measurements of glial fibrillary acidic protein, principal compound of intermediate filaments of astrocytes, and of phosphorylated neurofilament H and M epitopes in areas 10 and 24. All regions studied seem almost completely spared in normal old controls, with only the oldest ones exhibiting a weak percentage of beta-amyloid deposit and hardly any NFT. On the contrary, all AD and FAD cases were severely damaged as shown by statistically significant increased percentages of beta-amyloid deposit, as well as by a high number of NFT. FAD cases (all from the same family) had statistically more beta-amyloid and GFAP than sporadic AD cases in both areas 10 and 24 and statistically more NFT only in area 24. The correlation between the percentage of beta-amyloid and the number of NFT was significant only for area 24. Altogether, these data suggest that the frontal cortex can be spared by AD type lesions in normal aging, but is severely damaged in sporadic and still more in familial Alzheimer's disease. The frontal regions appear to be differentially vulnerable, with area 12 having the less amyloid burden, area 24 the less NFT and area 10 having both more amyloid and more NFT. This pattern of damage in frontal regions may represent a strong neuroanatomical support for the deterioration of attention and cognitive capacities as well as for the presence of emotional and behavioral troubles in AD patients.


Subject(s)
Aging/pathology , Alzheimer Disease/pathology , Frontal Lobe/pathology , Adult , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Female , Frontal Lobe/metabolism , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Male , Middle Aged , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology
15.
Brain Res Bull ; 80(4-5): 309-14, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19539730

ABSTRACT

The neuropathology of Alzheimer disease is characterized by senile plaques, neurofibrillary tangles and cell death. These hallmarks develop according to the differential vulnerability of brain networks, senile plaques accumulating preferentially in the associative cortical areas and neurofibrillary tangles in the entorhinal cortex and the hippocampus. We suggest that the main aetiological hypotheses such as the beta-amyloid cascade hypothesis or its variant, the synaptic beta-amyloid hypothesis, will have to consider neural networks not just as targets of degenerative processes but also as contributors of the disease's progression and of its phenotype. Three domains of research are highlighted in this review. First, the cerebral reserve and the redundancy of the network's elements are related to brain vulnerability. Indeed, an enriched environment appears to increase the cerebral reserve as well as the threshold of disease's onset. Second, disease's progression and memory performance cannot be explained by synaptic or neuronal loss only, but also by the presence of compensatory mechanisms, such as synaptic scaling, at the microcircuit level. Third, some phenotypes of Alzheimer disease, such as hallucinations, appear to be related to progressive dysfunction of neural networks as a result, for instance, of a decreased signal to noise ratio, involving a diminished activity of the cholinergic system. Overall, converging results from studies of biological as well as artificial neural networks lead to the conclusion that changes in neural networks contribute strongly to Alzheimer disease's progression.


Subject(s)
Alzheimer Disease/etiology , Brain/pathology , Nerve Net/pathology , Alzheimer Disease/pathology , Disease Progression , Humans , Models, Neurological , Neural Networks, Computer
16.
J Alzheimers Dis ; 15(1): 139-51, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18780974

ABSTRACT

We investigated how synaptic plasticity is related to the neurodegeneration process in the human dorsolateral prefrontal cortex. Pre- and postsynaptic proteins of Brodmann's area 9 from patients with Alzheimer's disease (AD) and age-matched controls were quantified by immunohistochemical methods and Western blots. The main finding was a significant increase in the expression of postsynaptic density protein PSD-95 in AD brains, revealed on both sections and immunoblots, while the expression of spinophilin, associated to spines, remained quantitatively unchanged despite qualitative changes with age and disease. Presynaptic protein alpha-synuclein indicated an increased immunohistochemical level, while synaptophysin remained unchanged. MAP2, a somatodendritic microtubule protein, as well as AD markers such as amyloid-beta protein and phosphorylated protein tau showed an increased expression on immunosections in AD. Altogether these changes suggest neuritic and synaptic reorganization in the process of AD. In particular, the significant increase in PSD-95 expression suggests a change in NMDA receptors trafficking and may represent a novel marker of functional significance for the disease.


Subject(s)
Alzheimer Disease/metabolism , Frontal Lobe/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Synapses/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Disks Large Homolog 4 Protein , Electrophoresis , Female , Humans , Immunohistochemistry , Male , Middle Aged
17.
Neurobiol Dis ; 30(3): 408-419, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18424056

ABSTRACT

In order to understand how plasticity is related to neurodegeneration, we studied synaptic proteins with quantitative immunohistochemistry in the entorhinal cortex from Alzheimer patients and age-matched controls. We observed a significant decrease in presynaptic synaptophysin and an increase in postsynaptic density protein PSD-95, positively correlated with beta amyloid and phosphorylated Tau proteins in Alzheimer cases. Furthermore, Alzheimer-like neuritic retraction was generated in okadaic acid (OA) treated SH-SY5Y neuroblastoma cells with no decrease in PSD-95 expression. However, in a SH-SY5Y clone with decreased expression of transcription regulator LMO4 (as observed in Alzheimer's disease) and increased neuritic length, PSD-95 expression was enhanced but did not change with OA treatment. Therefore, increased PSD-95 immunoreactivity in the entorhinal cortex might result from compensatory mechanisms, as in the SH-SY5Y clone, whereas increased Alzheimer-like Tau phosphorylation is not related to PSD-95 expression, as suggested by the OA-treated cell models.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/biosynthesis , Neurites/physiology , Okadaic Acid/toxicity , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Disks Large Homolog 4 Protein , Female , Gene Expression Regulation/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Middle Aged , Neurites/drug effects , Neurites/pathology , Phosphorylation , Up-Regulation/genetics , tau Proteins/metabolism
18.
Brain Res Mol Brain Res ; 115(2): 93-103, 2003 Jul 23.
Article in English | MEDLINE | ID: mdl-12877980

ABSTRACT

LMO4 is a transcription regulator interacting with proteins involved, among else, in tumorigenesis. Its function in the nervous system, and particularly in the adult nervous system, has however still to be elucidated. We decided to modify its expression in a neuronal model, human SH-SY5Y neuroblastoma cells, by permanent transfection of sense or anti-sense Lmo4 cDNAs. Generated clones overexpressing the Lmo4 transcript in sense orientation tended to aggregate. They showed significantly reduced average number of neurites per cell and average neuritic length per cell. The opposite was observed with clones overexpressing the anti-sense Lmo4 transcript. Furthermore, selected clones were subjected to 72 h long-term treatments with retinoic acid and phorbol ester (TPA), two biochemicals known to stimulate differentiation of non-transfected SH-SY5Y cells and other neuroblastoma cells. Neuritogenesis occurred after retinoic acid stimulation in all cases. The inhibitory effect of sense Lmo4 RNA overexpression on neuritic outgrowth was indeed prevented. The protein kinase C activator TPA could not induce neuritogenesis in SH-SY5Y cells overexpressing sense Lmo4 RNA. Thus, sense Lmo4 RNA overexpression, not Lmo4 endogenous transcription, overrides the stimulatory effect of TPA upon neuritic outgrowth. We also showed that Lmo4-dependent neuritic retraction and outgrowth correspond to altered phosphorylation of cytoskeletal proteins. Overall, Lmo4 RNA overexpression interferes with neuritic outgrowth, whereas anti-sense Lmo4 RNA expression favors neuritogenesis in SH-SY5Y cells. Consequently, changes in Lmo4 RNA expression levels might alter the rate of neuritic outgrowth in the developing and adult nervous system.


Subject(s)
Homeodomain Proteins/physiology , Neurites/physiology , RNA Precursors/physiology , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Blotting, Western/methods , Carcinogens/pharmacology , Cell Aggregation/drug effects , Cell Aggregation/genetics , Cell Aggregation/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cloning, Molecular , Cytoskeletal Proteins/metabolism , Gene Expression/physiology , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins , Models, Neurological , Neurites/drug effects , Neuroblastoma/pathology , Phorbol Esters/pharmacology , RNA, Antisense/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transfection/methods , Tretinoin/pharmacology , Tumor Cells, Cultured
19.
Neurobiol Dis ; 12(1): 46-55, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12609488

ABSTRACT

A Swiss frontotemporal dementia (FTD) kindred with extrapyramidal-like features and without motor neuron disease shows a brain pathology with ubiquitin-positive but tau-negative inclusions. Tau and neurofilament modifications are now studied here in three recently deceased family members. No major and specific decrease of tau was observed as described by others in, e.g., sporadic cases of FTD with absence of tau-positive inclusions. However, a slight decrease of tau, neurofilament, and synaptic proteins, resulting from frontal atrophy was detected. In parallel, polymorphic markers on chromosome 17q21-22, the centromeric region of chromosome 3 and chromosome 9, were tested. Haplotype analysis showed several recombination events for chromosomes 3 and 17, but patients shared a haplotype on chromosome 9q21-22. However as one of the patients exhibited Alzheimer and vascular dementia pathology with uncertain concomitant FTD, this locus is questionable. Altogether, these data indicate principally that the Swiss kindred is unlinked to locus 17q21-22, and that tau is not at the origin of FTD in this family.


Subject(s)
Cerebral Cortex/metabolism , Chromosomes, Human, Pair 17/genetics , Dementia/genetics , Dementia/metabolism , Neurofilament Proteins/deficiency , Neurons/metabolism , tau Proteins/deficiency , Adolescent , Adult , Aged , Aged, 80 and over , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Child , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 9/genetics , Dementia/pathology , Female , Genetic Linkage/genetics , Genetic Markers/genetics , Haplotypes/genetics , Humans , Immunohistochemistry , Infant, Newborn , Male , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/pathology , Pedigree , Switzerland , Synaptosomal-Associated Protein 25 , tau Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...