Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Biol Chem ; 292(30): 12713-12724, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28623233

ABSTRACT

Alzheimer disease is associated with deposition of the amyloidogenic peptide Aß in the brain. Passive immunization using Aß-specific antibodies has been demonstrated to reduce amyloid deposition both in vitro and in vivo Because N-terminally truncated pyroglutamate (pE)-modified Aß species (AßpE3) exhibit enhanced aggregation potential and propensity to form toxic oligomers, they represent particularly attractive targets for antibody therapy. Here we present three separate monoclonal antibodies that specifically recognize AßpE3 with affinities of 1-10 nm and inhibit AßpE3 fibril formation in vitro. In vivo application of one of these resulted in improved memory in AßpE3 oligomer-treated mice. Crystal structures of Fab-AßpE3 complexes revealed two distinct binding modes for the peptide. Juxtaposition of pyroglutamate pE3 and the F4 side chain (the "pEF head") confers a pronounced bulky hydrophobic nature to the AßpE3 N terminus that might explain the enhanced aggregation properties of the modified peptide. The deep burial of the pEF head by two of the antibodies explains their high target specificity and low cross-reactivity, making them promising candidates for the development of clinical antibodies.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Immunotherapy , Pyrrolidonecarboxylic Acid/immunology , Amyloid beta-Peptides/chemistry , Animals , Cells, Cultured , Mice
2.
Acta Neuropathol ; 126(2): 189-205, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23685882

ABSTRACT

N-truncated Aß4-42 is highly abundant in Alzheimer disease (AD) brain and was the first Aß peptide discovered in AD plaques. However, a possible role in AD aetiology has largely been neglected. In the present report, we demonstrate that Aß4-42 rapidly forms aggregates possessing a high aggregation propensity in terms of monomer consumption and oligomer formation. Short-term treatment of primary cortical neurons indicated that Aß4-42 is as toxic as pyroglutamate Aß3-42 and Aß1-42. In line with these findings, treatment of wildtype mice using intraventricular Aß injection induced significant working memory deficits with Aß4-42, pyroglutamate Aß3-42 and Aß1-42. Transgenic mice expressing Aß4-42 (Tg4-42 transgenic line) developed a massive CA1 pyramidal neuron loss in the hippocampus. The hippocampus-specific expression of Aß4-42 correlates well with age-dependent spatial reference memory deficits assessed by the Morris water maze test. Our findings indicate that N-truncated Aß4-42 triggers acute and long-lasting behavioral deficits comparable to AD typical memory dysfunction.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Nerve Degeneration/pathology , Neurons/drug effects , Alzheimer Disease/etiology , Amyloid/chemistry , Amyloid/ultrastructure , Amyloid beta-Peptides/chemistry , Animals , Behavior, Animal/physiology , Female , Fetus/cytology , Humans , Injections, Intraventricular , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Molecular Weight , Nerve Degeneration/etiology , Neurons/cytology , Neurons/pathology , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar , Solubility , Space Perception/physiology
3.
J Neurochem ; 123(4): 467-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22909011

ABSTRACT

Brain lipid homeostasis is important for maintenance of brain cell function and synaptic communications, and is intimately linked to age-related cognitive decline. Because of the blood-brain barrier's limiting nature, this tissue relies on a complex system for the synthesis and receptor-mediated uptake of lipids between the different networks of neurons and glial cells. Using immunofluorescence, we describe the region-specific expression of the lipolysis-stimulated lipoprotein receptor (LSR), in the mouse hippocampus, cerebellum Purkinje cells, the ependymal cell interface between brain parenchyma and cerebrospinal fluid, and the choroid plexus. Colocalization with cell-specific markers revealed that LSR was expressed in neurons, but not astrocytes. Latency in arms of the Y-maze exhibited by young heterozygote LSR(+/-) mice was significantly different as compared to control LSR(+/+), and increased in older LSR(+/-) mice. Filipin and Nile red staining revealed membrane cholesterol content accumulation accompanied by significantly altered distribution of LSR in the membrane, and decreased intracellular lipid droplets in the cerebellum and hippocampus of old LSR(+/-) mice, as compared to control littermates as well as young LSR(+/-) animals. These data therefore suggest a potential role of LSR in brain cholesterol distribution, which is particularly important in preserving neuronal integrity and thereby cognitive functions during aging.


Subject(s)
Aging/metabolism , Brain/cytology , Brain/metabolism , Cholesterol/metabolism , Neurons/metabolism , Receptors, LDL/metabolism , Age Factors , Animals , Choroid Plexus/metabolism , Filipin/metabolism , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphopyruvate Hydratase/metabolism , Receptors, LDL/deficiency
4.
J Neurosci ; 30(22): 7516-27, 2010 Jun 02.
Article in English | MEDLINE | ID: mdl-20519526

ABSTRACT

The development of novel therapeutic strategies for Alzheimer's disease (AD) represents one of the biggest unmet medical needs today. Application of neurotrophic factors able to modulate neuronal survival and synaptic connectivity is a promising therapeutic approach for AD. We aimed to determine whether the loco-regional delivery of ciliary neurotrophic factor (CNTF) could prevent amyloid-beta (Abeta) oligomer-induced synaptic damages and associated cognitive impairments that typify AD. To ensure long-term administration of CNTF in the brain, we used recombinant cells secreting CNTF encapsulated in alginate polymers. The implantation of these bioreactors in the brain of Abeta oligomer-infused mice led to a continuous secretion of recombinant CNTF and was associated with the robust improvement of cognitive performances. Most importantly, CNTF led to full recovery of cognitive functions associated with the stabilization of synaptic protein levels in the Tg2576 AD mouse model. In vitro as well as in vivo, CNTF activated a Janus kinase/signal transducer and activator of transcription-mediated survival pathway that prevented synaptic and neuronal degeneration. These preclinical studies suggest that CNTF and/or CNTF receptor-associated pathways may have AD-modifying activity through protection against progressive Abeta-related memory deficits. Our data also encourage additional exploration of ex vivo gene transfer for the prevention and/or treatment of AD.


Subject(s)
Alzheimer Disease/complications , Ciliary Neurotrophic Factor/biosynthesis , Ciliary Neurotrophic Factor/therapeutic use , Memory Disorders/etiology , Memory Disorders/therapy , Synapses/drug effects , Alzheimer Disease/genetics , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/genetics , Brain/pathology , Cell Count/methods , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Ciliary Neurotrophic Factor/administration & dosage , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Synapses/metabolism , Synaptosomes/metabolism , Synaptosomes/pathology , Synaptosomes/ultrastructure , Time Factors , Transfection/methods
5.
Biochim Biophys Acta ; 1801(8): 791-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20211757

ABSTRACT

Alzheimer's disease (AD) is a major public health concern due to longer life expectancy in the Western countries. Amyloid-beta (Abeta) oligomers are considered the proximate effectors in the early stages of AD. AD-related cognitive impairment, synaptic loss and neurodegeneration result from interactions of Abeta oligomers with the synaptic membrane and subsequent activation of pro-apoptotic signalling pathways. Therefore, membrane structure and lipid status appear determinant in Abeta-induced toxicity. Numerous epidemiological studies have highlighted the beneficial influence of docosahexaenoic acid (DHA, C22:6 n-3) on the preservation of synaptic function and memory capacities in aged individuals or upon Abeta exposure, whereas its deficiency is presented as a risk factor for AD. An elevated number of studies have been reporting the beneficial effects of dietary DHA supplementation on cognition and synaptic integrity in various AD models. In this review, we describe the important potential of DHA to preserve neuronal and brain functions and classified its numerous molecular and cellular effects from impact on membrane lipid content and organisation to activation of signalling pathways sustaining synaptic function and neuronal survival. DHA appears as one of the most valuable diet ingredients whose neuroprotective properties could be crucial for designing nutrition-based strategies able to prevent AD as well as other lipid- and age-related diseases whose prevalence is progressing in elderly populations.


Subject(s)
Alzheimer Disease/pathology , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Mice , Synapses/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Cognition Disorders/prevention & control , Cytoprotection/drug effects , Docosahexaenoic Acids/metabolism , Humans , Models, Biological , Neurons/drug effects , Neuroprotective Agents/pharmacology
6.
FASEB J ; 24(11): 4218-28, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20647547

ABSTRACT

As a hepatic receptor for triglyceride-rich lipoproteins, the lipolysis-stimulated lipoprotein receptor (LSR) may be involved in the dynamics of lipid distribution between the liver and peripheral tissues. Here, we explore the potential role of leptin in regulating LSR. At physiological concentrations (1-10 ng/ml), leptin increased LSR protein and mRNA levels in Hepa1-6 cells through an ERK1/2-dependent and α-amanitin-sensitive pathway. In vivo, leptin treatment of C57BL6/Rj mice (1 µg 2×/d, 8 d) led to a significant increase in hepatic LSR mRNA and protein, decreased liver triglycerides and increased VLDL secretion as compared to controls. LSR(+/-) mice with elevated postprandial lipemia placed on a high-fat (60% kcal) diet exhibited accelerated weight gain and increased fat mass as compared to controls. While plasma leptin levels were increased 3-fold, hepatic leptin receptor protein levels and phosphorylation of ERK1/2 were significantly reduced. Therefore, leptin is an important regulator of LSR protein levels providing the means for the control of hepatic uptake of lipids during the postprandial phase. However, this may no longer be functional in LSR(+/-) mice placed under a chronic dietary fat load, suggesting that this animal model could be useful for the study of molecular mechanisms involved in peripheral leptin resistance.


Subject(s)
Leptin/pharmacology , Lipid Metabolism/drug effects , Lipolysis/drug effects , Liver/drug effects , Postprandial Period , Receptors, Lipoprotein/metabolism , Up-Regulation/drug effects , Animals , Blotting, Western , Body Weight/drug effects , Cell Line , Fluorescent Antibody Technique , Leptin/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction
7.
Mol Neurobiol ; 55(7): 5594-5610, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28983842

ABSTRACT

Amyloid precursor protein (APP) is cleaved not only to generate the amyloid peptide (Aß), involved in neurodegenerative processes, but can also be metabolized by alpha secretase to produce and release soluble N-terminal APP (sAPPα), which has many properties including the induction of axonal elongation and neuroprotection. The mechanisms underlying the properties of sAPPα are not known. Here, we used proteomic analysis of mouse cortico-hippocampal membranes to identify the neuronal specific alpha3 (α3)-subunit of the plasma membrane enzyme Na, K-ATPase (NKA) as a new binding partner of sAPPα. We showed that sAPPα recruits very rapidly clusters of α3-NKA at neuronal surface, and its binding triggers a cascade of events promoting sAPPα-induced axonal outgrowth. The binding of sAPPα with α3-NKA was not observed for sAPPα-induced Aß1-42 oligomers neuroprotection, neither the downstream events particularly the interaction of sAPPα with APP before endocytosis, ERK signaling, and the translocation of SET from the nucleus to the plasma membrane. These data suggest that the mechanisms of the axonal growth promoting and neuroprotective properties of sAPPα appear to be specific and independent. The signals at the cell surface specific to trigger these mechanisms require further study.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Axons/metabolism , Neuroprotection , Peptide Fragments/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Membrane/metabolism , DNA-Binding Proteins , Endocytosis , Histone Chaperones , Humans , MAP Kinase Signaling System , Mice, Inbred C57BL , Models, Biological , Neurites/metabolism , Oncogene Proteins/metabolism , Protein Binding , Solubility
8.
J Alzheimers Dis ; 62(1): 213-226, 2018.
Article in English | MEDLINE | ID: mdl-29439327

ABSTRACT

BACKGROUND: With current treatments for Alzheimer's disease (AD) only providing temporary symptomatic benefits, disease modifying drugs are urgently required. This approach relies on improved understanding of the early pathophysiology of AD. A new hypothesis has emerged, in which early memory loss is considered a synapse failure caused by soluble amyloid-ß oligomers (Aßo). These small soluble Aßo, which precede the formation of larger fibrillar assemblies, may be the main cause of early AD pathologies. OBJECTIVE: The aim of the current study was to investigate the effect of acute administration of stabilized low-n amyloid-ß1-42 oligomers (Aßo1-42) on cognitive, inflammatory, synaptic, and neuronal markers in the rat. METHODS: Female and male Lister Hooded rats received acute intracerebroventricular (ICV) administration of either vehicle or 5 nmol of Aßo1-42 (10µL). Cognition was assessed in the novel object recognition (NOR) paradigm at different time points. Levels of inflammatory (IL-1ß, IL-6, TNF-α), synaptic (PSD-95, SNAP-25), and neuronal (n-acetylaspartate, parvalbumin-positive cells) markers were investigated in different brain regions (prefrontal and frontal cortex, striatum, dorsal and ventral hippocampus). RESULTS: Acute ICV administration of Aßo1-42 induced robust and enduring NOR deficits. These deficits were reversed by acute administration of donepezil and rolipram but not risperidone. Postmortem analysis revealed an increase in inflammatory markers, a decrease in synaptic markers and parvalbumin containing interneurons in the frontal cortex, with no evidence of widespread neuronal loss. CONCLUSION: Taken together the results suggest that acute administration of soluble low-n Aßo may be a useful model to study the early mechanisms involved in AD and provide us with a platform for testing novel therapeutic approaches that target the early underlying synaptic pathology.


Subject(s)
Amyloid beta-Peptides/metabolism , Cognition/physiology , Inflammation/metabolism , Memory Disorders/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition/drug effects , Disease Models, Animal , Donepezil/pharmacology , Female , Inflammation/drug therapy , Inflammation/pathology , Male , Memory Disorders/drug therapy , Memory Disorders/pathology , Neurons/drug effects , Neurons/pathology , Nootropic Agents/pharmacology , Random Allocation , Rats , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Risperidone/pharmacology , Rolipram/pharmacology , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
9.
J Neurol Sci ; 262(1-2): 27-36, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17681547

ABSTRACT

Alzheimer's disease (AD) is a major public health concern in all countries. Although the precise cause of AD is still unknown, a growing body of evidence supports the notion that soluble amyloid beta-peptide (Abeta) may be the proximate cause of synaptic injuries and neuronal death early in the disease. AD patients display lower levels of docosahexaenoic acid (DHA, C22:6 ; n-3) in plasma and brain tissues as compared to age-matched controls. Furthermore, epidemiological studies suggest that high DHA intake might have protective properties against neurodegenerative diseases. These observations are supported by in vivo studies showing that DHA-rich diets limits the synaptic loss and cognitive defects induced by Abeta peptide. Although the molecular basis of these neuroprotective effects remains unknown, several mechanisms have been proposed such as (i) regulation of the expression of potentially protective genes, (ii) activation of anti-inflammatory pathways, (iii) modulation of functional properties of the synaptic membranes along with changes in their physicochemical and structural features.


Subject(s)
Alzheimer Disease/diet therapy , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/antagonists & inhibitors , Brain/drug effects , Brain/metabolism , Docosahexaenoic Acids/therapeutic use , Encephalitis/diet therapy , Neuroprotective Agents/therapeutic use , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Brain/physiopathology , Docosahexaenoic Acids/metabolism , Encephalitis/metabolism , Encephalitis/physiopathology , Food, Formulated/standards , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Neuroprotective Agents/metabolism , Synapses/drug effects , Synapses/metabolism
10.
Alzheimers Res Ther ; 9(1): 69, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28851448

ABSTRACT

BACKGROUND: Polyunsaturated fatty acids play a crucial role in neuronal function, and the modification of these compounds in the brain could have an impact on neurodegenerative diseases such as Alzheimer's disease. Despite the fact that arachidonic acid is the second foremost polyunsaturated fatty acid besides docosahexaenoic acid, its role and the regulation of its transfer and mobilization in the brain are poorly known. METHODS: Two groups of 39 adult male BALB/c mice were fed with an arachidonic acid-enriched diet or an oleic acid-enriched diet, respectively, for 12 weeks. After 10 weeks on the diet, mice received intracerebroventricular injections of either NaCl solution or amyloid-ß peptide (Aß) oligomers. Y-maze and Morris water maze tests were used to evaluate short- and long-term memory. At 12 weeks on the diet, mice were killed, and blood, liver, and brain samples were collected for lipid and protein analyses. RESULTS: We found that the administration of an arachidonic acid-enriched diet for 12 weeks induced short-term memory impairment and increased deleterious effects of Aß oligomers on learning abilities. These cognitive alterations were associated with modifications of expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors, postsynaptic density protein 95, and glial fibrillary acidic protein in mouse cortex or hippocampus by the arachidonic acid-enriched diet and Aß oligomer administration. This diet also led to an imbalance between the main ω-6 fatty acids and the ω-3 fatty acids in favor of the first one in erythrocytes and the liver as well as in the hippocampal and cortical brain structures. In the cortex, the dietary arachidonic acid also induced an increase of arachidonic acid-containing phospholipid species in phosphatidylserine class, whereas intracerebroventricular injections modified several arachidonic acid- and docosahexaenoic acid-containing species in the four phospholipid classes. Finally, we observed that dietary arachidonic acid decreased the expression of the neuronal form of acyl-coenzyme A synthetase 4 in the hippocampus and increased the cytosolic phospholipase A2 activation level in the cortices of the mice. CONCLUSIONS: Dietary arachidonic acid could amplify Aß oligomer neurotoxicity. Its consumption could constitute a risk factor for Alzheimer's disease in humans and should be taken into account in future preventive strategies. Its deleterious effect on cognitive capacity could be linked to the balance between arachidonic acid-mobilizing enzymes.


Subject(s)
Amyloid beta-Peptides/toxicity , Arachidonic Acid/adverse effects , Diet/adverse effects , Maze Learning/physiology , Peptide Fragments/toxicity , Receptors, AMPA/metabolism , Animals , Arachidonic Acid/administration & dosage , Astrocytes/metabolism , Cell Proliferation/physiology , Cerebral Cortex/metabolism , Coenzyme A Ligases/metabolism , Cytosol/metabolism , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Male , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice, Inbred BALB C , Neurons/metabolism , Oleic Acid/administration & dosage , Phospholipases A2/metabolism , Risk Factors
11.
FASEB J ; 19(1): 85-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15486059

ABSTRACT

Recent data have revealed that soluble oligomeric forms of amyloid peptide (Abeta) may be the proximate effectors of the neuronal injury and death occurring in Alzheimer's disease (AD). However, the molecular mechanisms associated with the neuronal cell death induced by the nonfibrillar Abeta remain to be elucidated. In this study, we investigated the role of the cytosolic Ca2+-dependent phospholipase A2 (cPLA2), and its associated metabolic pathway, i.e., the arachidonic acid (AA) cascade, in the apoptotic cell death induced by soluble oligomers of Abeta. The treatment of rat cortical neurons with low concentrations of soluble Abeta(1-40) or Abeta(1-42) peptide resulted in an early calcium-dependent release of AA associated with a transient relocalization of cPLA2. Both cPLA2 antisense oligonucleotides and a selective inhibitor of cPLA2 activity abolished the release of AA from neurons and also protected cells against apoptosis induced by Abeta. Furthermore, inhibitors of the PKC, p38, and MEK/ERK pathways that are involved in cPLA2 phosphorylation and activation reduced Abeta-induced cell death. Finally, we demonstrate that inhibitors of cyclooxygenase-2 reduced the Abeta-induced cell death by 55%. Our studies suggest a novel neuronal response of soluble oligomers of Abeta, which occurs through a cPLA2 signaling cascade and an AA-dependent death pathway. This may prove to be crucial in AD processes and could provide important targets for drug development.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis/physiology , Cytosol/enzymology , Neurons/enzymology , Neurons/physiology , Peptide Fragments/metabolism , Phospholipases A/metabolism , Animals , Arachidonic Acid/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cyclooxygenase 2 , Humans , Membrane Proteins , Mitogen-Activated Protein Kinases/metabolism , Phospholipases A2 , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Solubility
12.
Nat Commun ; 7: 12504, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27509875

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenase-1 (COX-1) and COX-2 enzymes. The NLRP3 inflammasome is a multi-protein complex responsible for the processing of the proinflammatory cytokine interleukin-1ß and is implicated in many inflammatory diseases. Here we show that several clinically approved and widely used NSAIDs of the fenamate class are effective and selective inhibitors of the NLRP3 inflammasome via inhibition of the volume-regulated anion channel in macrophages, independently of COX enzymes. Flufenamic acid and mefenamic acid are efficacious in NLRP3-dependent rodent models of inflammation in air pouch and peritoneum. We also show therapeutic effects of fenamates using a model of amyloid beta induced memory loss and a transgenic mouse model of Alzheimer's disease. These data suggest that fenamate NSAIDs could be repurposed as NLRP3 inflammasome inhibitors and Alzheimer's disease therapeutics.


Subject(s)
Alzheimer Disease/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flufenamic Acid/pharmacology , Inflammasomes/metabolism , Mefenamic Acid/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Alzheimer Disease/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Death , Chloride Channels/metabolism , Cysteine/metabolism , Female , Genotype , Inflammation , Interleukin-1beta/metabolism , Macrophages/metabolism , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pattern Recognition, Visual/drug effects , Rats
13.
J Neurosci ; 23(2): 462-9, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12533606

ABSTRACT

We recently demonstrated that the 118-135 putative transmembrane domain of prion protein (PrP) exhibited membrane fusogenic properties and induced apoptotic neuronal cell death of rat cortical neurons, independently of its aggregation state. The aim of the present study was to analyze the in vivo neurotoxicity of the prion fragment P118-135 and to evaluate the potential role of the physiological isoform of PrP in the P118-135-induced cell death. Here, we demonstrate that the nonfibrillar P118-135 is cytotoxic to retinal neurons in vivo as monitored by intravitreal inoculation and recording of the electrical activity of retina and tissue examination. Moreover, knock-out PrP gene mice exhibit similar sensitivity to the nonfibrillar P118-135-induced cell death and electrical perturbations, strongly suggesting that cell death occurs independently of PrP expression. Interestingly, a variant nonfusogenic P118-135 peptide (termed P118-135theta) had no effects on in vivo neuronal viability, suggesting that the P118-135-induced cell death is mediated by its membrane destabilizing properties. These data have further been confirmed in vitro. We show that the fusogenic peptide P118-135 induces death of cultured neurons from both wild-type and knock-out PrP gene mice via an apoptotic-mediated pathway, involving early caspase activation and DNA fragmentation. Altogether these results emphasize the neurotoxicity of the fusogenic nonfibrillar PrP transmembrane domain and indicate that fibril formation and PrP expression are not obligatory requirements for neuronal cell death. The use of synthetic prion peptides could provide insights into the understanding of neuronal loss mechanisms that take place during the development of the various types of spongiform encephalopathies.


Subject(s)
Peptide Fragments/toxicity , Prions/biosynthesis , Prions/toxicity , Animals , Apoptosis , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , DNA Fragmentation , Drug Administration Routes , Electroretinography/drug effects , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Membrane Fusion/physiology , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Peptide Fragments/administration & dosage , Prion Diseases/etiology , Prions/administration & dosage , Prions/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary/physiology , Retina/cytology , Retina/drug effects
14.
FASEB J ; 18(7): 836-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15001562

ABSTRACT

Neuronal cell death in Alzheimer's disease (AD) is partly induced by the interaction of the amyloid-beta peptide (Abeta) with the plasma membrane of target cells. Accordingly, recent studies have suggested that cholesterol, an important component of membranes that controls their physical properties and functions, plays a critical role in neurodegenerative diseases. We report here that the enrichment of the neuronal plasma membrane with cholesterol protects cortical neurons from apoptosis induced by soluble oligomers of the Abeta(1-40) peptide. Conversely, cholesterol depletion using cyclodextrin renders cells more vulnerable to the cytotoxic effects of the Abeta-soluble oligomers. Increasing the cholesterol content of small unilamellar liposomes also decreases Abeta-dependent liposome fusion. We clearly demonstrate that cholesterol protection is specific to the soluble conformation of Abeta, because we observed no protective effects on cortical neurons treated by amyloid fibrils of the Abeta(1-40) peptide. This may provide a new opportunity for the development of an effective AD therapy as well as elucidate the impact of the cholesterol level during AD development.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Apoptosis/drug effects , Cholesterol/pharmacology , Membrane Lipids/pharmacology , Neurons/drug effects , Peptide Fragments/antagonists & inhibitors , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Biopolymers , Caspase 8 , Caspases/metabolism , Cells, Cultured/drug effects , Cells, Cultured/ultrastructure , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Lipid Bilayers , Liposomes , Membrane Fluidity , Membrane Fusion/drug effects , Neurons/ultrastructure , Oxidative Stress , Peptide Fragments/pharmacology , Peptide Fragments/toxicity , Protein Conformation , Rats , Rats, Wistar , Solubility
15.
Sci Rep ; 5: 17338, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626428

ABSTRACT

Full-length Aß1-42 and Aß1-40, N-truncated pyroglutamate Aß3-42 and Aß4-42 are major variants in the Alzheimer brain. Aß4-42 has not been considered as a therapeutic target yet. We demonstrate that the antibody NT4X and its Fab fragment reacting with both the free N-terminus of Aß4-x and pyroglutamate Aß3-X mitigated neuron loss in Tg4-42 mice expressing Aß4-42 and completely rescued spatial reference memory deficits after passive immunization. NT4X and its Fab fragment also rescued working memory deficits in wild type mice induced by intraventricular injection of Aß4-42. NT4X reduced pyroglutamate Aß3-x, Aßx-40 and Thioflavin-S positive plaque load after passive immunization of 5XFAD mice. Aß1-x and Aßx-42 plaque deposits were unchanged. Importantly, for the first time, we demonstrate that passive immunization using the antibody NT4X is therapeutically beneficial in Alzheimer mouse models showing that N-truncated Aß starting with position four in addition to pyroglutamate Aß3-x is a relevant target to fight Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Immunization, Passive/methods , Peptide Fragments/immunology , Alzheimer Disease/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Disease Models, Animal , Humans , Mice , Rats
16.
J Alzheimers Dis ; 45(1): 195-204, 2015.
Article in English | MEDLINE | ID: mdl-25690661

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease that has been linked to changes in cholesterol metabolism. Neuronal cholesterol content significantly influences the pro-apoptotic effect of amyloid-ß peptide42 (Aß42), which plays a key role in AD development. We previously reported that aged mice with reduced expression of the lipolysis stimulated lipoprotein receptor (LSR+/-), demonstrate membrane cholesterol accumulation and decreased intracellular lipid droplets in several brain regions, suggesting a potential role of LSR in brain cholesterol distribution. We questioned if these changes rendered the LSR+/- mouse more susceptible to Aß42-induced cognitive and biochemical changes. Results revealed that intracerebroventricular injection of oligomeric Aß42 in male 15-month old LSR+/+ and LSR+/- mice led to impairment in learning and long-term memory and decreased cortical cholesterol content of both groups; these effects were significantly amplified in the Aß42-injected LSR+/- group. Total latency of the Morris test was significantly and negatively correlated with cortical cholesterol content of the LSR+/- mice, but not of controls. Significantly lower cortical PSD95 and SNAP-25 levels were detected in Aß42-injected LSR+/- mice as compared to Aß42-injected LSR+/+ mice. In addition, 24S-hydroxy cholesterol metabolite levels were significantly higher in the cortex of LSR+/- mice. Taken together, these results suggest that changes in cortex cholesterol regulation as a result of the LSR+/- genotype were linked to increased susceptibility to amyloid stress, and we would therefore propose the aged LSR+/- mouse as a new model for understanding the link between modified cholesterol regulation as a risk factor for AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholesterol/metabolism , Peptide Fragments/pharmacology , Receptors, Lipoprotein/deficiency , Analysis of Variance , Animals , Disks Large Homolog 4 Protein , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Guanylate Kinases/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydroxycholesterols/metabolism , Maze Learning/drug effects , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger , Receptors, Lipoprotein/genetics , Regression Analysis , Synaptosomal-Associated Protein 25/metabolism
17.
Brain Res Brain Res Protoc ; 11(1): 46-51, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12697262

ABSTRACT

Membrane perturbing effects have been described in neurodegenerative process like Alzheimer's disease and prion disorders. For example, non fibrillar amyloid-beta peptides (Abeta) implicated in Alzheimer's disease may exert its toxicity via membrane perturbation. Membrane organisation can be evaluated by its influence on lateral diffusion of lipids, which itself can be measured by FRAP (fluorescence recovery after photobleaching). We used this technique to study the effects of Abeta on membrane fluidity (Pillot et al., manuscript in preparation). We propose here a simple adaptation of FRAP using standard confocal laser scanning microscopy (CLSM). As a test experiment, we analysed the lateral diffusion of a fluorescent analogue of sphingomyelin and were able to demonstrate its increase upon cholesterol depletion induced by methyl-beta-cyclodextrin (cdx).


Subject(s)
4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Brain/pathology , Cell Membrane/pathology , Fluorescence Recovery After Photobleaching/methods , Membrane Fluidity/physiology , Membrane Lipids/metabolism , Neurodegenerative Diseases/pathology , Neurons/pathology , beta-Cyclodextrins , Brain/metabolism , Brain/physiopathology , Cell Membrane/metabolism , Cholesterol/deficiency , Cyclodextrins/pharmacology , Fluorescence Recovery After Photobleaching/instrumentation , Humans , Microscopy, Confocal , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Sphingomyelins , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/physiology
18.
Acta Neuropathol Commun ; 1: 56, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24252153

ABSTRACT

BACKGROUND: The amyloid hypothesis in Alzheimer disease (AD) considers amyloid ß peptide (Aß) deposition causative in triggering down-stream events like neurofibrillary tangles, cell loss, vascular damage and memory decline. In the past years N-truncated Aß peptides especially N-truncated pyroglutamate AßpE3-42 have been extensively studied. Together with full-length Aß1-42 and Aß1-40, N-truncated AßpE3-42 and Aß4-42 are major variants in AD brain. Although Aß4-42 has been known for a much longer time, there is a lack of studies addressing the question whether AßpE3-42 or Aß4-42 may precede the other in Alzheimer's disease pathology. RESULTS: Using different Aß antibodies specific for the different N-termini of N-truncated Aß, we discovered that Aß4-x preceded AßpE3-x intraneuronal accumulation in a transgenic mouse model for AD prior to plaque formation. The novel Aß4-x immunoreactive antibody NT4X-167 detected high molecular weight aggregates derived from N-truncated Aß species. While NT4X-167 significantly rescued Aß4-42 toxicity in vitro no beneficial effect was observed against Aß1-42 or AßpE3-42 toxicity. Phenylalanine at position four of Aß was imperative for antibody binding, because its replacement with alanine or proline completely prevented binding. Although amyloid plaques were observed using NT4X-167 in 5XFAD transgenic mice, it barely reacted with plaques in the brain of sporadic AD patients and familial cases with the Arctic, Swedish and the presenilin-1 PS1Δ9 mutation. A consistent staining was observed in blood vessels in all AD cases with cerebral amyloid angiopathy. There was no cross-reactivity with other aggregates typical for other common neurodegenerative diseases showing that NT4X-167 staining is specific for AD. CONCLUSIONS: Aß4-x precedes AßpE3-x in the well accepted 5XFAD AD mouse model underlining the significance of N-truncated species in AD pathology. NT4X-167 therefore is the first antibody reacting with Aß4-x and represents a novel tool in Alzheimer research.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/administration & dosage , Neurons/physiology , Aged , Aged, 80 and over , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Animals , Brain/drug effects , Brain/physiopathology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Epitope Mapping , Female , Humans , Male , Mice, Transgenic , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Plaque, Amyloid/drug therapy , Plaque, Amyloid/physiopathology , Rats, Wistar
19.
Neurobiol Aging ; 33(6): 1123.e17-29, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22188721

ABSTRACT

Soluble beta-amyloid (Aß) oligomers are considered to putatively play a critical role in the early synapse loss and cognitive impairment observed in Alzheimer's disease. We previously demonstrated that Aß oligomers activate cytosolic phospholipase A(2) (cPLA(2)), which specifically releases arachidonic acid from membrane phospholipids. We here observed that cPLA(2) gene inactivation prevented the alterations of cognitive abilities and the reduction of hippocampal synaptic markers levels noticed upon a single intracerebroventricular injection of Aß oligomers in wild type mice. We further demonstrated that the Aß oligomer-induced sphingomyelinase activation was suppressed and that phosphorylation of Akt/protein kinase B (PKB) was preserved in neuronal cells isolated from cPLA(2)(-/-) mice. Interestingly, expression of the Aß precursor protein (APP) was reduced in hippocampus homogenates and neuronal cells from cPLA(2)(-/-) mice, but the relationship with the resistance of these mice to the Aß oligomer toxicity requires further investigation. These results therefore show that cPLA(2) plays a key role in the Aß oligomer-associated neurodegeneration, and as such represents a potential therapeutic target for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Memory Disorders/enzymology , Memory Disorders/genetics , Neurodegenerative Diseases/enzymology , Peptide Fragments/toxicity , Phospholipases A2, Cytosolic/physiology , Animals , Cells, Cultured , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/chemically induced , Mice , Mice, Inbred BALB C , Mice, Knockout , Neurodegenerative Diseases/chemically induced
20.
Biochimie ; 91(6): 804-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19303044

ABSTRACT

In the absence of efficient diagnostic and therapeutic tools, Alzheimer's disease (AD) is a major public health concern due to longer life expectancy in the Western countries. Although the precise cause of AD is still unknown, soluble beta-amyloid (Abeta) oligomers are considered the proximate effectors of the synaptic injury and neuronal death occurring in the early stages of AD. Abeta oligomers may directly interact with the synaptic membrane, leading to impairment of synaptic functions and subsequent signalling pathways triggering neurodegeneration. Therefore, membrane structure and lipid status should be considered determinant factors in Abeta-oligomer-induced synaptic and cell injuries, and therefore AD progression. Numerous epidemiological studies have highlighted close relationships between AD incidence and dietary patterns. Among the nutritional factors involved, lipids significantly influence AD pathogenesis. It is likely that maintenance of adequate membrane lipid content could prevent the production of Abeta peptide as well as its deleterious effects upon its interaction with synaptic membrane, thereby protecting neurons from Abeta-induced neurodegeneration. As major constituents of neuronal lipids, n-3 polyunsaturated fatty acids are of particular interest in the prevention of AD valuable diet ingredients whose neuroprotective properties could be essential for designing preventive nutrition-based strategies. In this review, we discuss the functional relevance of neuronal membrane features with respect to susceptibility to Abeta oligomers and AD pathogenesis, as well as the prospective capacities of lipids to prevent or to delay the disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Lipid Metabolism/physiology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Apoptosis/physiology , Fatty Acids, Omega-3/metabolism , Humans , Membrane Microdomains/metabolism , Models, Biological , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL