Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
ACS Chem Neurosci ; 6(4): 559-69, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25611616

ABSTRACT

The amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD). Preventing deregulated APP processing by inhibiting amyloidogenic processing of carboxy-terminal fragments (APP-CTFs), and reducing the toxic effect of amyloid beta (Aß) peptides remain an effective therapeutic strategy. We report the design of piperazine-containing compounds derived from chloroquine structure and evaluation of their effects on APP metabolism and ability to modulate the processing of APP-CTF and the production of Aß peptide. Compounds which retained alkaline properties and high affinity for acidic cell compartments were the most effective. The present study demonstrates that (1) the amino side chain of chloroquine can be efficiently substituted by a bis(alkylamino)piperazine chain, (2) the quinoline nucleus can be replaced by a benzyl or a benzimidazole moiety, and (3) pharmacomodulation of the chemical structure allows the redirection of APP metabolism toward a decrease of Aß peptide release, and increased stability of APP-CTFs and amyloid intracellular fragment. Moreover, the benzimidazole compound 29 increases APP-CTFs in vivo and shows promising activity by the oral route. Together, this family of compounds retains a lysosomotropic activity which inhibits lysosome-related Aß production, and is likely to be beneficial for therapeutic applications in AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Chloroquine/analogs & derivatives , Neuroprotective Agents/chemistry , Quinolines/chemistry , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Chloroquine/chemistry , Chloroquine/pharmacology , Drug Design , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Protein Stability/drug effects , Quinolines/pharmacology , Water/chemistry
2.
Lancet Neurol ; 9(11): 1118-27, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20934914

ABSTRACT

Alzheimer's disease (AD) is classically defined as a dual clinicopathological entity. The recent advances in use of reliable biomarkers of AD that provide in-vivo evidence of the disease has stimulated the development of new research criteria that reconceptualise the diagnosis around both a specific pattern of cognitive changes and structural/biological evidence of Alzheimer's pathology. This new diagnostic framework has stimulated debate about the definition of AD and related conditions. The potential for drugs to intercede in the pathogenic cascade of the disease adds some urgency to this debate. This paper by the International Working Group for New Research Criteria for the Diagnosis of AD aims to advance the scientific discussion by providing broader diagnostic coverage of the AD clinical spectrum and by proposing a common lexicon as a point of reference for the clinical and research communities. The cornerstone of this lexicon is to consider AD solely as a clinical and symptomatic entity that encompasses both predementia and dementia phases.


Subject(s)
Alzheimer Disease/classification , Alzheimer Disease/diagnosis , Alzheimer Disease/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Chicago , Dementia/cerebrospinal fluid , Dementia/classification , Dementia/diagnosis , Humans
3.
Mol Ther ; 18(1): 44-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19654569

ABSTRACT

The development of Alzheimer's disease (AD) is closely connected with cholesterol metabolism. Cholesterol increases the production and deposition of amyloid-beta (Abeta) peptides that result in the formation of amyloid plaques, a hallmark of the pathology. In the brain, cholesterol is synthesized in situ but cannot be degraded nor cross the blood-brain barrier. The major exportable form of brain cholesterol is 24S-hydroxycholesterol, an oxysterol generated by the neuronal cholesterol 24-hydroxylase encoded by the CYP46A1 gene. We report that the injection of adeno-associated vector (AAV) encoding CYP46A1 in the cortex and hippocampus of APP23 mice before the onset of amyloid deposits markedly reduces Abeta peptides, amyloid deposits and trimeric oligomers at 12 months of age. The Morris water maze (MWM) procedure also demonstrated improvement of spatial memory at 6 months, before the onset of amyloid deposits. AAV5-wtCYP46A1 vector injection in the cortex and hippocampus of amyloid precursor protein/presenilin 1 (APP/PS) mice after the onset of amyloid deposits also reduced markedly the number of amyloid plaques in the hippocampus, and to a less extent in the cortex, 3 months after the injection. Our data demonstrate that neuronal overexpression of CYP46A1 before or after the onset of amyloid plaques significantly reduces Abeta pathology in mouse models of AD.


Subject(s)
Alzheimer Disease/therapy , Amyloid/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Steroid Hydroxylases/physiology , Alzheimer Disease/metabolism , Animals , Blotting, Western , Cell Line , Cholesterol 24-Hydroxylase , Enzyme-Linked Immunosorbent Assay , Humans , Hydroxycholesterols/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Steroid Hydroxylases/genetics
4.
Science ; 323(5916): 946-51, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19213921

ABSTRACT

Deposition of the amyloid-beta peptide is a pathological hallmark of Alzheimer's disease. A high-throughput functional genomics screen identified G protein-coupled receptor 3 (GPR3), a constitutively active orphan G protein-coupled receptor, as a modulator of amyloid-beta production. Overexpression of GPR3 stimulated amyloid-beta production, whereas genetic ablation of GPR3 prevented accumulation of the amyloid-beta peptide in vitro and in an Alzheimer's disease mouse model. GPR3 expression led to increased formation and cell-surface localization of the mature gamma-secretase complex in the absence of an effect on Notch processing. GPR3 is highly expressed in areas of the normal human brain implicated in Alzheimer's disease and is elevated in the sporadic Alzheimer's disease brain. Thus, GPR3 represents a potential therapeutic target for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Adult , Aged , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , Humans , Male , Mice , Middle Aged , Protein Structure, Tertiary , Receptors, Notch/metabolism , Signal Transduction
5.
J Biol Chem ; 284(18): 12447-58, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19240038

ABSTRACT

Clathrin-dependent endocytosis is mediated by a tightly regulated network of molecular interactions that provides essential protein-protein and protein-lipid binding activities. Here we report the hydrolysis of the alpha- and beta2-subunits of the tetrameric adaptor protein complex 2 by calpain. Calcium-dependent alpha- and beta2-adaptin hydrolysis was observed in several rat tissues, including brain and primary neuronal cultures. Neuronal alpha- and beta2-adaptin cleavage was inducible by glutamate stimulation and was accompanied by the decreased endocytosis of transferrin. Heterologous expression of truncated forms of the beta2-adaptin subunit significantly decreased the membrane recruitment of clathrin and inhibited clathrin-mediated receptor endocytosis. Moreover, the presence of truncated beta2-adaptin sensitized neurons to glutamate receptor-mediated excitotoxicity. Proteolysis of alpha- and beta2-adaptins, as well as the accessory clathrin adaptors epsin 1, adaptor protein 180, and the clathrin assembly lymphoid myeloid leukemia protein, was detected in brain tissues after experimentally induced ischemia and in cases of human Alzheimer disease. The present study further clarifies the central role of calpain in regulating clathrin-dependent endocytosis and provides evidence for a novel mechanism through which calpain activation may promote neurodegeneration: the sensitization of cells to glutamate-mediated excitotoxicity via the decreased internalization of surface receptors.


Subject(s)
Adaptor Protein Complex alpha Subunits/metabolism , Adaptor Protein Complex beta Subunits/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Calpain/metabolism , Clathrin/metabolism , Endocytosis , Neurons/metabolism , Adaptor Protein Complex alpha Subunits/genetics , Adaptor Protein Complex beta Subunits/genetics , Adaptor Proteins, Vesicular Transport , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Calcium/metabolism , Calpain/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Clathrin/genetics , Female , Glutamic Acid/metabolism , Humans , Hydrolysis , Male , Membrane Lipids/genetics , Membrane Lipids/metabolism , Monomeric Clathrin Assembly Proteins/genetics , Monomeric Clathrin Assembly Proteins/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Rats, Wistar
7.
Neurobiol Dis ; 33(3): 422-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19110058

ABSTRACT

Gene dosage effects of Amyloid precursor protein (APP) can cause familial AD. Recent evidence suggest that microRNA (miRNA) pathways, implicated in gene transcriptional control, could be involved in the development of sporadic Alzheimer's disease (AD). We therefore investigated whether miRNAs could participate in the regulation of APP gene expression. We show that miRNAs belonging to the miR-20a family (that is, miR-20a, miR-17-5p and miR-106b) could regulate APP expression in vitro and at the endogenous level in neuronal cell lines. A tight correlation between these miRNAs and APP was found during brain development and in differentiating neurons. We thus identify miRNAs as novel endogenous regulators of APP expression, suggesting that variations in miRNA expression could contribute to changes in APP expression in the brain during development and disease. This possibility is further corroborated by the observation that a statistically significant decrease in miR-106b expression was found in sporadic AD patients.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Gene Expression Regulation , MicroRNAs/physiology , Neurons/physiology , Receptors, Cell Surface/genetics , Actins/metabolism , Animals , Blotting, Northern , Blotting, Western , Brain/embryology , Brain/growth & development , Brain/physiology , Cell Line , Cells, Cultured , Densitometry , Humans , Mice , Neurogenesis , Protease Nexins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
8.
J Alzheimers Dis ; 14(4): 437-40, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18688095

ABSTRACT

Most dementing disorders result from a degenerating process named tauopathy. Alzheimer disease is the most frequent one, but only one among the large spectrum of tau-related diseases. Cognitive impairment is related, first of all, to the neocortical location of this degenerating process. However, the nature and the mechanisms leading to tauopathy can be very different. This is demonstrated by familial mutations on the tau gene as well as by the different morphological and biochemical patterns of tau lesions. Therefore there is no doubt that tau is an etiological agent. But the persistent and unsolved question is the basic mechanism leading to neurodegeneration: is it due to the toxic effect of aggregated tau, or a loss of tau function, or both? Some answers may come from a more focused interest towards sporadic tauopathies. Most of them are characterized by a degenerating process starting in a specific and vulnerable brain area and consuming the connected neuronal network, like a chain reaction. In other words, sporadic tauopathies are mostly a destabilization of specific neuronal networks that should be modeled for an efficient therapeutic approach.


Subject(s)
Nerve Degeneration/pathology , tau Proteins/physiology , Alzheimer Disease/pathology , Animals , Humans , Neurodegenerative Diseases/pathology , tau Proteins/chemistry
10.
Expert Rev Proteomics ; 5(2): 207-24, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18466052

ABSTRACT

Microtubule-associated Tau proteins belong to a family of factors that polymerize tubulin dimers and stabilize microtubules. Tau is strongly expressed in neurons, localized in the axon and is essential for neuronal plasticity and network. From the very beginning of Tau discovery, proteomics methods have been essential to the knowledge of Tau biochemistry and biology. In this review, we have summarized the main contributions of several proteomic methods in the understanding of Tau, including expression, post-translational modifications and structure, in both physiological and pathophysiological aspects. Finally, recent advances in proteomics technology are essential to develop further therapeutic targets and early predictive and discriminative diagnostic assays for Alzheimer's disease and related disorders.


Subject(s)
Alzheimer Disease/etiology , Nervous System Diseases/etiology , tau Proteins/genetics , Alzheimer Disease/diagnosis , Humans , Nervous System Diseases/diagnosis , Protein Isoforms , Protein Processing, Post-Translational , tau Proteins/chemistry , tau Proteins/physiology
11.
Proc Natl Acad Sci U S A ; 105(17): 6415-20, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18434550

ABSTRACT

Although the role of APP and PSEN genes in genetic Alzheimer's disease (AD) cases is well established, fairly little is known about the molecular mechanisms affecting Abeta generation in sporadic AD. Deficiency in Abeta clearance is certainly a possibility, but increased expression of proteins like APP or BACE1/beta-secretase may also be associated with the disease. We therefore investigated changes in microRNA (miRNA) expression profiles of sporadic AD patients and found that several miRNAs potentially involved in the regulation of APP and BACE1 expression appeared to be decreased in diseased brain. We show here that miR-29a, -29b-1, and -9 can regulate BACE1 expression in vitro. The miR-29a/b-1 cluster was significantly (and AD-dementia-specific) decreased in AD patients displaying abnormally high BACE1 protein. Similar correlations between expression of this cluster and BACE1 were found during brain development and in primary neuronal cultures. Finally, we provide evidence for a potential causal relationship between miR-29a/b-1 expression and Abeta generation in a cell culture model. We propose that loss of specific miRNAs can contribute to increased BACE1 and Abeta levels in sporadic AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , MicroRNAs/genetics , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Brain/pathology , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Mice , Up-Regulation/genetics
12.
Neurobiol Aging ; 29(11): 1619-30, 2008 Nov.
Article in English | MEDLINE | ID: mdl-17531353

ABSTRACT

Several lines of evidence suggest that the glutamatergic system is severely impaired in Alzheimer disease (AD). Here, we assessed the status of glutamatergic terminals in AD using the first available specific markers, the vesicular glutamate transporters VGLUT1 and VGLUT2. We quantified VGLUT1 and VGLUT2 in the prefrontal dorsolateral cortex (Brodmann area 9) of controls and AD patients using specific antiserums. A dramatic decrease in VGLUT1 and VGLUT2 was observed in AD using Western blot. Similar decreases were observed in an independent group of subjects using immunoautoradiography. The VGLUT1 reduction was highly correlated with the degree of cognitive impairment, assessed with the clinical dementia rating (CDR) score. A significant albeit weaker correlation was also observed with VGLUT2. These findings provide evidence indicating that glutamatergic systems are severely impaired in the A9 region of AD patients and that this impairment is strongly correlated with the progression of cognitive decline. Our results suggest that VGLUT1 expression in the prefrontal cortex could be used as a valuable neurochemical marker of dementia in AD.


Subject(s)
Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Prefrontal Cortex/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/complications , Cognition Disorders/complications , Female , Humans , Male , Middle Aged , Statistics as Topic
13.
Neurodegener Dis ; 4(6): 413-23, 2007.
Article in English | MEDLINE | ID: mdl-17934324

ABSTRACT

BACKGROUND/AIMS: Alzheimer's disease (AD) is characterized by extracellular Abeta peptide deposition originating from amyloid precursor protein cleavage and intracellular neurofibrillary tangles resulting from pathological tau protein aggregation. These processes are accompanied by dramatic neuronal losses, further leading to different cognitive impairments. Neuronal death signalings involve gene expression modifications that rely on transcription factor alterations. Herein, we investigated the fate of the Sp family of transcription factors in postmortem brains from patients with AD disease and in different contexts of neuronal death. METHODS/RESULTS: By immunohistochemistry we found that the Sp3 and Sp4 levels were dramatically increased and associated with neurofibrillary tangles and pathological tau presence in neurons from the CA1 region of the hippocampus, as well as the entorhinal cortex of AD patient brains. The Sp transcription factor expression levels were further analyzed in cortical neurons in which death is induced by amyloid precursor protein signaling targeting. While the Sp1 levels remained constant, the Sp4 levels were slightly upregulated in response to the death signal. The Sp3 isoforms were rather degraded. Interestingly, when overexpressed by transfection experiments, the three Sp family members induced neuronal apoptosis, Sp3 and Sp4 being the most potent proapoptotic factors over Sp1. CONCLUSION: Our data evidence Sp3 and Sp4 as new hallmarks of AD in postmortem human brains and further point out that Sp proteins are potential triggers of neuronal death signaling cascades.


Subject(s)
Alzheimer Disease/pathology , Brain/metabolism , Gene Expression Regulation/physiology , Sp3 Transcription Factor/metabolism , Sp4 Transcription Factor/metabolism , Amyloid beta-Protein Precursor/immunology , Animals , Animals, Newborn , Antibodies/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Case-Control Studies , Cells, Cultured , Cerebellum/cytology , Drug Interactions , Humans , Leupeptins/pharmacology , Mice , Neurons/drug effects , Neurons/physiology , Oligopeptides/pharmacology , Postmortem Changes , Time Factors , Transfection/methods , tau Proteins/metabolism
14.
Lancet Neurol ; 6(8): 734-46, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17616482

ABSTRACT

The NINCDS-ADRDA and the DSM-IV-TR criteria for Alzheimer's disease (AD) are the prevailing diagnostic standards in research; however, they have now fallen behind the unprecedented growth of scientific knowledge. Distinctive and reliable biomarkers of AD are now available through structural MRI, molecular neuroimaging with PET, and cerebrospinal fluid analyses. This progress provides the impetus for our proposal of revised diagnostic criteria for AD. Our framework was developed to capture both the earliest stages, before full-blown dementia, as well as the full spectrum of the illness. These new criteria are centred on a clinical core of early and significant episodic memory impairment. They stipulate that there must also be at least one or more abnormal biomarkers among structural neuroimaging with MRI, molecular neuroimaging with PET, and cerebrospinal fluid analysis of amyloid beta or tau proteins. The timeliness of these criteria is highlighted by the many drugs in development that are directed at changing pathogenesis, particularly at the production and clearance of amyloid beta as well as at the hyperphosphorylation state of tau. Validation studies in existing and prospective cohorts are needed to advance these criteria and optimise their sensitivity, specificity, and accuracy.


Subject(s)
Alzheimer Disease/diagnosis , Diagnostic and Statistical Manual of Mental Disorders , Severity of Illness Index , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/complications , Amyloid beta-Peptides/cerebrospinal fluid , Diagnostic Imaging/methods , Disease Progression , Humans , Memory Disorders/diagnosis , Memory Disorders/etiology , National Institutes of Health (U.S.)/standards , Time Factors , United States , tau Proteins/cerebrospinal fluid
15.
Neurobiol Dis ; 27(2): 164-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17566751

ABSTRACT

Alzheimer's disease (AD) is the most common form of neurodegenerative disorder in the ageing population. It is characterized by the cerebral accumulation of toxic amyloid-beta peptide assemblies (Abeta). The serine protease plasmin, which is generated from the inactive zymogen plasminogen through its proteolytic cleavage by tissue- (tPA) or urokinase-type plasminogen activator, has been implicated in the catabolism of Abeta peptides. In this report, we studied the regulation of tPA activity in vivo during ageing in normal mice and in a mouse model of AD characterized by an exacerbated endogenous Abeta accumulation. We observed that cerebral tPA activity was decreased during ageing in normal mice and that this effect was worsened in mice overproducing Abeta peptides. These phenomena result, respectively, from a decrease in tPA expression and from an increase in the production of one of the tPA inhibitors, the plasminogen activator inhibitor type 1 (PAI-1). A similar study in sporadic AD and age-matched control brain tissues revealed that the tPA proteolytic activity was negatively correlated to Abeta peptides levels supporting the data observed in mice. Altogether, our data support a model in which amyloid deposition induces a decrease in tPA activity through the overproduction of PAI-1 by activated glial cells.


Subject(s)
Aging/physiology , Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Brain/enzymology , Tissue Plasminogen Activator/metabolism , Animals , Brain/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , Mice, Transgenic , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Int J Med Sci ; 4(3): 140-5, 2007 May 12.
Article in English | MEDLINE | ID: mdl-17505559

ABSTRACT

In Alzheimer disease, neuronal degeneration and the presence of neurofibrillary tangles correlate with the severity of cognitive decline. Neurofibrillary tangles contain the antigenic profile of many cell cycle markers, reflecting a re-entry into the cell cycle by affected neurons. However, while such a cell cycle re-entry phenotype is an early and consistent feature of Alzheimer disease, the mechanisms responsible for neuronal cell cycle are unclear. In this regard, given that a dysregulated cell cycle is a characteristic of cancer, we speculated that alterations in oncogenic proteins may play a role in neurodegeneration. To this end, in this study, we examined brain tissue from cases of Alzheimer disease for the presence of BRCA1, a known regulator of cell cycle, and found intense and specific localization of BRCA1 to neurofibrillary tangles, a hallmark lesion of the disease. Analysis of clinically normal aged brain tissue revealed systematically less BRCA1, and surprisingly in many cases with apparent phosphorylated tau-positive neurofibrillary tangles, BRCA1 was absent, yet BRCA1 was present in all cases of Alzheimer disease. These findings not only further define the cell cycle reentry phenotype in Alzheimer disease but also indicate that the neurofibrillary tangles which define Alzheimer disease may have a different genesis from the neurofibrillary tangles of normal aging.


Subject(s)
Alzheimer Disease/physiopathology , BRCA1 Protein/physiology , Cell Cycle/physiology , Neurons/physiology , Aged , Aged, 80 and over , Aging/physiology , BRCA1 Protein/analysis , Humans , Middle Aged , Neurofibrillary Tangles/chemistry
17.
Proc Natl Acad Sci U S A ; 104(19): 8167-72, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470798

ABSTRACT

The amyloid precursor protein (APP) undergoes sequential cleavages to generate various polypeptides, including the amyloid-beta protein (Abeta), which forms amyloid plaques in Alzheimer's disease (AD), secreted APPalpha (sAPPalpha) which enhances memory, and the APP intracellular domain (AICD), which has been implicated in the regulation of gene transcription and calcium signaling. The beta-site APP cleaving enzyme 1 (BACE1) cleaves APP in an activity-dependent manner to form Abeta, AICD, and secreted APPbeta. Because this neural activity was shown to diminish synaptic transmission in vitro [Kamenetz F, Tomita T, Hsieh H, Seabrook G, Borchelt D, Iwatsubo T, Sisodia S, Malinow R (2003) Neuron 37:925-937], the prevailing notion has been that this pathway diminishes synaptic function. Here we investigated the role of this pathway in vivo. We studied transgenic mice overproducing APP that do not develop AD pathology or memory deficits but instead exhibit enhanced spatial memory. We showed enhanced synaptic plasticity in the hippocampus that depends on prior synaptic activity. We found that the enhanced memory and synaptic plasticity are abolished by the ablation of one or both copies of the BACE1 gene, leading to a significant decrease in AICD but not of any other APP cleavage products. In contrast to the previously described negative effect of BACE1-mediated cleavage of APP on synaptic function in vitro, our in vivo work indicates that BACE1-mediated cleavage of APP can facilitate learning, memory, and synaptic plasticity.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Protein Precursor/physiology , Aspartic Acid Endopeptidases/physiology , Memory , Neuronal Plasticity , Synapses/physiology , Amyloid beta-Protein Precursor/chemistry , Animals , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
J Biol Chem ; 282(25): 18197-18205, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17468104

ABSTRACT

Amyloid precursor protein (APP) metabolism is central to the pathogenesis of Alzheimer disease. We showed recently that the amyloid intracellular domain (AICD), which is released by gamma-secretase cleavage of APP C-terminal fragments (CTFs), is strongly increased in cells treated with alkalizing drugs (Vingtdeux, V., Hamdane, M., Bégard, S., Loyens, A., Delacourte, A., Beauvillain, J.-C., Buée, L., Marambaud, P., and Sergeant, N. (2007) Neurobiol. Dis. 25, 686-696). Herein, we aimed to determine the cell compartment in which AICD accumulates. We show that APP-CTFs and AICD are present in multivesicular structures. Multivesicular bodies contain intraluminal vesicles (known as exosomes) when released in the extracellular space. We demonstrate that APP, APP-CTFs, and AICD are integrated and secreted within exosomes in differentiated neuroblastoma and primary neuronal culture cells. Together with recent data showing that amyloid-beta is also found in exosomes, our data show that multivesicular bodies are essential organelles for APP metabolism and that all APP metabolites can be secreted in the extracellular space.


Subject(s)
Amyloid/metabolism , Enzyme Inhibitors/pharmacology , Animals , Brain/embryology , Cell Line, Tumor , Endosomes/metabolism , Humans , Macrolides/pharmacology , Models, Biological , Neuroblastoma/metabolism , Neurons/metabolism , Organelles/metabolism , Protein Structure, Tertiary , Rats , Rats, Wistar
19.
Arch Neurol ; 64(4): 583-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17420322

ABSTRACT

OBJECTIVE: To describe the neuropathological and biochemical findings of the brain examination of a patient enrolled in the AN-1792(QS-21) trial with an initial clinical diagnosis of Alzheimer disease (AD), in whom Lewy body variant was thereafter clinically diagnosed. DESIGN: A case report. SETTING: University memory clinic. Patient A 74-year-old woman with clinical features of probable AD. Intervention The patient received 2 injections of 225 mug of AN-1792 (beta-amyloid [Abeta]) plus 50 mug of the adjuvant QS-21 at an interval of 4 weeks. The patient was an antibody responder with an IgG anti-AN-1792 antibody titer exceeding 10 000 and an IgM titer exceeding 3500. Maximum serum anti-Abeta titers were reached in 4.7 months. During the 3 following years, while the Mini-Mental State Examination score remained globally stable despite several confusional episodes, she developed clinical features of dementia with Lewy bodies. The patient died 34 months postimmunization. An autopsy was performed. MAIN OUTCOME MEASURES: Neuropathological and biochemical examination of the brain using standardized evaluation for tau, beta-amyloid, and synuclein deposits. RESULTS: Neither neuropathological nor biochemical examinations showed amyloid deposit in the brain of this immunized patient. For tau deposition, Braak stage was IV/VI, and the Western blot analysis score was 9c/10. The neuropathological semiquantitative score for alpha-synuclein aggregation was 4. There was no inflammation. These results were compared with those of an age-matched patient with AD and a control devoid of any neurological disease. CONCLUSION: In this Lewy body variant case, with globally stable functional and cognitive features, Abeta immunization resulted in a significant clearance of amyloid deposits, with remaining tau and synuclein pathological features in the brain. Patients with a Lewy body variant of AD should not be excluded from enrollment in Abeta-immunization trials.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Lewy Body Disease/immunology , Aged , Alzheimer Disease/blood , Alzheimer Disease/prevention & control , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/metabolism , Brain/metabolism , Brain/pathology , Fatal Outcome , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Lewy Body Disease/metabolism
20.
Neurobiol Dis ; 25(3): 686-96, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17207630

ABSTRACT

The amyloid precursor protein (APP) metabolism is central to pathogenesis of Alzheimer's disease (AD). Parenchymal amyloid deposits, a neuropathological hallmark of AD, are composed of amyloid-beta peptides (Abeta). Abeta derives from the amyloid precursor protein (APP) by sequential cleavages by beta- and gamma-secretases. Gamma-secretase cleavage releases the APP intracellular domain (AICD), suggested to mediate a nuclear signaling. Physiologically, AICD is seldom detected and thus supposed to be rapidly degraded. The mechanisms responsible of its degradation remain unknown. We used a pharmacological approach and showed that several alkalizing drugs induce the accumulation of AICD in neuroblastoma SY5Y cell lines stably expressing APP constructs. Moreover, alkalizing drugs induce AICD accumulation in naive SY5Y, HEK and COS cells. This accumulation is not mediated by the proteasome or metallopeptidases and is not the result of an increased gamma-secretase activity since the gamma-secretase cleavage of Notch1 and N-Cadherin is not affected by alkalizing drug treatments. Altogether, our data demonstrate for the first time that alkalizing drugs induce the accumulation of AICD, a mechanism likely mediated by the endosome/lysosome pathway.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Hydrogen-Ion Concentration , Alkalies/metabolism , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , COS Cells , Cadherins/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration/drug effects , Kidney/cytology , Lysosomes/metabolism , Macrolides/pharmacology , Neuroblastoma , Protein Structure, Tertiary , Receptors, Notch/metabolism , Solubility , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...