Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Aging Neurosci ; 14: 845291, 2022.
Article in English | MEDLINE | ID: mdl-35572125

ABSTRACT

The APOE gene is diversified by three alleles ε2, ε3, and ε4 encoding corresponding apolipoprotein (apo) E isoforms. Possession of the ε4 allele is signified by increased risks of age-related cognitive decline, Alzheimer's disease (AD), and the rate of AD dementia progression. ApoE is secreted by astrocytes as high-density lipoprotein-like particles and these are internalized by neurons upon binding to neuron-expressed apoE receptors. ApoE isoforms differentially engage neuronal plasticity through poorly understood mechanisms. We examined here the effects of native apoE lipoproteins produced by immortalized astrocytes homozygous for ε2, ε3, and ε4 alleles on the maturation and the transcriptomic profile of primary hippocampal neurons. Control neurons were grown in the presence of conditioned media from Apoe -/- astrocytes. ApoE2 and apoE3 significantly increase the dendritic arbor branching, the combined neurite length, and the total arbor surface of the hippocampal neurons, while apoE4 fails to produce similar effects and even significantly reduces the combined neurite length compared to the control. ApoE lipoproteins show no systemic effect on dendritic spine density, yet apoE2 and apoE3 increase the mature spines fraction, while apoE4 increases the immature spine fraction. This is associated with opposing effects of apoE2 or apoE3 and apoE4 on the expression of NR1 NMDA receptor subunit and PSD95. There are 1,062 genes differentially expressed across neurons cultured in the presence of apoE lipoproteins compared to the control. KEGG enrichment and gene ontology analyses show apoE2 and apoE3 commonly activate expression of genes involved in neurite branching, and synaptic signaling. In contrast, apoE4 cultured neurons show upregulation of genes related to the glycolipid metabolism, which are involved in dendritic spine turnover, and those which are usually silent in neurons and are related to cell cycle and DNA repair. In conclusion, our work reveals that lipoprotein particles comprised of various apoE isoforms differentially regulate various neuronal arbor characteristics through interaction with neuronal transcriptome. ApoE4 produces a functionally distinct transcriptomic profile, which is associated with attenuated neuronal development. Differential regulation of neuronal transcriptome by apoE isoforms is a newly identified biological mechanism, which has both implication in the development and aging of the CNS.

2.
Acta Neuropathol Commun ; 9(1): 157, 2021 09 26.
Article in English | MEDLINE | ID: mdl-34565486

ABSTRACT

Prion diseases or prionoses are a group of rapidly progressing and invariably fatal neurodegenerative diseases. The pathogenesis of prionoses is associated with self-replication and connectomal spread of PrPSc, a disease specific conformer of the prion protein. Microglia undergo activation early in the course of prion pathogenesis and exert opposing roles in PrPSc mediated neurodegeneration. While clearance of PrPSc and apoptotic neurons have disease-limiting effect, microglia-driven neuroinflammation bears deleterious consequences to neuronal networks. Apolipoprotein (apo) E is a lipid transporting protein with pleiotropic functions, which include controlling of the phagocytic and inflammatory characteristics of activated microglia in neurodegenerative diseases. Despite the significance of microglia in prion pathogenesis, the role of apoE in prionoses has not been established. We showed here that infection of wild type mice with 22L mouse adapted scrapie strain is associated with significant increase in the total brain apoE protein and mRNA levels and also with a conspicuous cell-type shift in the apoE expression. There is reduced expression of apoE in activated astrocytes and marked upregulation of apoE expression by activated microglia. We also showed apoE ablation exaggerates PrPSc mediated neurodegeneration. Apoe-/- mice have shorter disease incubation period, increased load of spongiform lesion, pronounced neuronal loss, and exaggerated astro and microgliosis. Astrocytes of Apoe-/- mice display salient upregulation of transcriptomic markers defining A1 neurotoxic astrocytes while microglia show upregulation of transcriptomic markers characteristic for microglial neurodegenerative phenotype. There is impaired clearance of PrPSc and dying neurons by microglia in Apoe-/- mice along with increased level of proinflammatory cytokines. Our work indicates that apoE absence renders clearance of PrPSc and dying neurons by microglia inefficient, while the excess of neuronal debris promotes microglial neurodegenerative phenotype aggravating the vicious cycle of neuronal death and neuroinflammation.


Subject(s)
Apolipoproteins E/metabolism , Microglia/pathology , Nerve Degeneration/pathology , Prion Diseases/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nerve Degeneration/metabolism , Phenotype , Prion Diseases/metabolism
3.
Mol Neurodegener ; 15(1): 50, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32907613

ABSTRACT

BACKGROUND: Disruption of ß-amyloid (Aß) homeostasis is the initial culprit in Alzheimer's disease (AD) pathogenesis. Astrocytes respond to emerging Aß plaques by altering their phenotype and function, yet molecular mechanisms governing astrocytic response and their precise role in countering Aß deposition remain ill-defined. Peroxiredoxin (PRDX) 6 is an enzymatic protein with independent glutathione peroxidase (Gpx) and phospholipase A2 (PLA2) activities involved in repair of oxidatively damaged cell membrane lipids and cellular signaling. In the CNS, PRDX6 is uniquely expressed by astrocytes and its exact function remains unexplored. METHODS: APPswe/PS1dE9 AD transgenic mice were once crossed to mice overexpressing wild-type Prdx6 allele or to Prdx6 knock out mice. Aß pathology and associated neuritic degeneration were assessed in mice aged 10 months. Laser scanning confocal microscopy was used to characterize Aß plaque morphology and activation of plaque-associated astrocytes and microglia. Effect of Prdx6 gene dose on plaque seeding was assessed in mice aged six months. RESULTS: We show that hemizygous knock in of the overexpressing Prdx6 transgene in APPswe/PS1dE9 AD transgenic mice promotes selective enticement of astrocytes to Aß plaques and penetration of plaques by astrocytic processes along with increased number and phagocytic activation of periplaque microglia. This effects suppression of nascent plaque seeding and remodeling of mature plaques consequently curtailing brain Aß load and Aß-associated neuritic degeneration. Conversely, Prdx6 haplodeficiency attenuates astro- and microglia activation around Aß plaques promoting Aß deposition and neuritic degeneration. CONCLUSIONS: We identify here PRDX6 as an important factor regulating response of astrocytes toward Aß plaques. Demonstration that phagocytic activation of periplaque microglia vary directly with astrocytic PRDX6 expression level implies previously unappreciated astrocyte-guided microglia effect in Aß proteostasis. Our showing that upregulation of PRDX6 attenuates Aß pathology may be of therapeutic relevance for AD.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Peroxiredoxin VI/metabolism , Plaque, Amyloid/metabolism , Proteostasis/physiology , Amyloid beta-Peptides/metabolism , Animals , Brain/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/pathology
4.
Mol Neurobiol ; 56(3): 2073-2091, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29987703

ABSTRACT

PrPSc is an infectious and disease-specific conformer of the prion protein, which accumulation in the CNS underlies the pathology of prion diseases. PrPSc replicates by binding to the cellular conformer of the prion protein (PrPC) expressed by host cells and rendering its secondary structure a likeness of itself. PrPC is a plasma membrane anchored protein, which constitutively recirculates between the cell surface and the endocytic compartment. Since PrPSc engages PrPC along this trafficking pathway, its replication process is often referred to as "recycling propagation." Certain monoclonal antibodies (mAbs) directed against prion protein can abrogate the presence of PrPSc from prion-infected cells. However, the precise mechanism(s) underlying their therapeutic propensities remains obscure. Using N2A murine neuroblastoma cell line stably infected with 22L mouse-adapted scrapie strain (N2A/22L), we investigated here the modus operandi of the 6D11 clone, which was raised against the PrPSc conformer and has been shown to permanently clear prion-infected cells from PrPSc presence. We determined that 6D11 mAb engages and sequesters PrPC and PrPSc at the cell surface. PrPC/6D11 and PrPSc/6D11 complexes are then endocytosed from the plasma membrane and are directed to lysosomes, therefore precluding recirculation of nascent PrPSc back to the cell surface. Targeting PrPSc by 6D11 mAb to the lysosomal compartment facilitates its proteolysis and eventually shifts the balance between PrPSc formation and degradation. Ongoing translation of PrPC allows maintaining the steady-state level of prion protein within the cells, which was not depleted under 6D11 mAb treatment. Our findings demonstrate that through disrupting recycling propagation of PrPSc and promoting its degradation, 6D11 mAb restores cellular proteostasis of prion protein.


Subject(s)
Antibodies, Monoclonal , Lysosomes/metabolism , PrPSc Proteins/metabolism , Prion Diseases/metabolism , Prion Proteins/metabolism , Animals , Cell Line, Tumor , Mice , Proteolysis , Proteostasis , Scrapie/metabolism
5.
Oncotarget ; 8(25): 39941-39942, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28537920
6.
Mol Neurodegener ; 12(1): 12, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28143566

ABSTRACT

BACKGROUND: APOE genotype is the foremost genetic factor modulating ß-amyloid (Aß) deposition and risk of sporadic Alzheimer's disease (AD). Here we investigated how APOE genotype influences response to anti-Aß immunotherapy. METHODS: APPSW/PS1dE9 (APP) transgenic mice with targeted replacement of the murine Apoe gene for human APOE alleles received 10D5 anti-Aß or TY11-15 isotype control antibodies between the ages of 12 and 15 months. RESULTS: Anti-Aß immunization decreased both the load of fibrillar plaques and the load of Aß immunopositive plaques in mice of all APOE backgrounds. Although the relative reduction in parenchymal Aß plaque load was comparable across all APOE genotypes, APP/ε4 mice showed the greatest reduction in the absolute Aß plaque load values, given their highest baseline. The immunization stimulated phagocytic activation of microglia, which magnitude adjusted for the post-treatment plaque load was the greatest in APP/ε4 mice implying association between the ε4 allele and impaired Aß phagocytosis. Perivascular hemosiderin deposits reflecting ensued microhemorrhages were associated with vascular Aß (VAß) and ubiquitously present in control mice of all APOE genotypes, although in APP/ε3 mice their incidence was the lowest. Anti-Aß immunization significantly reduced VAß burden but increased the number of hemosiderin deposits across all APOE genotypes with the strongest and the weakest effect in APP/ε2 and APP/ε3 mice, respectively. CONCLUSIONS: Our studies indicate that APOE genotype differentially modulates microglia activation and Aß plaque load reduction during anti-Aß immunotherapy. The APOE ε3 allele shows strong protective effect against immunotherapy associated microhemorrhages; while, conversely, the APOE ε2 allele increases risk thereof.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Apolipoproteins E/genetics , Animals , Disease Models, Animal , Genotype , Humans , Immunization, Passive , Immunohistochemistry , Mice , Mice, Transgenic , Plaque, Amyloid/genetics
9.
Acta Neuropathol Commun ; 2: 75, 2014 Jun 28.
Article in English | MEDLINE | ID: mdl-24972680

ABSTRACT

Accumulation of ß-amyloid (Aß) in the brain is essential to Alzheimer's disease (AD) pathogenesis. Carriers of the apolipoprotein E (APOE) ε4 allele demonstrate greatly increased AD risk and enhanced brain Aß deposition. In contrast, APOE ε2 allele carries show reduced AD risk, later age of disease onset, and lesser Aß accumulation. However, it remains elusive whether the apoE2 isoform exerts truly protective effect against Aß pathology or apoE2 plays deleterious role albeit less pronounced than the apoE4 isoform. Here, we characterized APPSW/PS1dE9/APOE ε2-TR (APP/E2) and APPSW/PS1dE9/APOE ε4-TR (APP/E4) mice, with targeted replacement (TR) of the murine Apoe for human ε2 or ε4 alleles, and used these models to investigate effects of pharmacological inhibition of the apoE/Aß interaction on Aß deposition and neuritic degeneration. APP/E2 and APP/E4 mice replicate differential effect of human apoE isoforms on Aß pathology with APP/E4 mice showing a several-fold greater load of Aß plaques, insoluble brain Aß levels, Aß oligomers, and density of neuritic plaques than APP/E2 mice. Furthermore, APP/E4 mice, but not APP/E2 mice, exhibit memory impairment on object recognition and radial arm maze tests. Between the age of 6 and 10 months APP/E2 and APP/E4 mice received treatment with Aß12-28P, a non-toxic, synthetic peptide homologous to the apoE binding motif within the Aß sequence, which competitively blocks the apoE/Aß interaction. In both lines, the treatment significantly reduced brain Aß accumulation, co-accumulation of apoE within Aß plaques, and neuritic degeneration, and prevented memory deficit in APP/E4 mice. These results indicate that both apoE2 and apoE4 isoforms contribute to Aß deposition and future therapies targeting the apoE/Aß interaction could produce favorable outcome in APOE ε2 and ε4 allele carriers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E2/metabolism , Apolipoprotein E4/metabolism , Brain/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/pathology , Disease Models, Animal , Female , Humans , Memory/physiology , Mice , Mice, Transgenic , Plaque, Amyloid/etiology , Plaque, Amyloid/metabolism
10.
Ann Neurol ; 75(5): 684-99, 2014 May.
Article in English | MEDLINE | ID: mdl-24687915

ABSTRACT

OBJECTIVE: Proteolytic cleavage of the amyloid precursor protein (APP) generates ß-amyloid (Aß) peptides. Prolonged accumulation of Aß in the brain underlies the pathogenesis of Alzheimer disease (AD) and is regarded as a principal target for development of disease-modifying therapeutics. METHODS: Using Chinese hamster ovary (CHO) APP751SW cells, we identified and characterized effects of 2-([pyridine-2-ylmethyl]-amino)-phenol (2-PMAP) on APP steady-state level and Aß production. Outcomes of 2-PMAP treatment on Aß accumulation and associated memory deficit were studied in APPSW /PS1dE9 AD transgenic model mice. RESULTS: In CHO APP751SW cells, 2-PMAP lowered the steady-state APP level and inhibited Aßx-40 and Aßx-42 production in a dose-response manner with a minimum effective concentration ≤ 0.5µM. The inhibitory effect of 2-PMAP on translational efficiency of APP mRNA into protein was directly confirmed using a 35S-methionine/cysteine metabolic labeling technique, whereas APP mRNA level remained unaltered. Administration of 2-PMAP to APPSW /PS1dE9 mice reduced brain levels of full-length APP and its C-terminal fragments and lowered levels of soluble Aßx-40 and Aßx-42 . Four-month chronic treatment of APPSW /PS1dE9 mice revealed no observable toxicity and improved animals' memory performance. 2-PMAP treatment also caused significant reduction in brain Aß deposition determined by both unbiased quantification of Aß plaque load and biochemical analysis of formic acid-extracted Aßx-40 and Aßx-42 levels and the level of oligomeric Aß. INTERPRETATION: We demonstrate the potential of modulating APP steady-state expression level as a safe and effective approach for reducing Aß deposition in AD transgenic model mice.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Gene Expression Regulation , Plaque, Amyloid/prevention & control , Amyloid beta-Protein Precursor/physiology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics
11.
Am J Pathol ; 182(5): 1750-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23499462

ABSTRACT

Accumulation of ß-amyloid (Aß) in the brain is a key event in Alzheimer disease pathogenesis. Apolipoprotein (Apo) E is a lipid carrier protein secreted by astrocytes, which shows inherent affinity for Aß and has been implicated in the receptor-mediated Aß uptake by neurons. To characterize ApoE involvement in the intraneuronal Aß accumulation and to investigate whether blocking the ApoE/Aß interaction could reduce intraneuronal Aß buildup, we used a noncontact neuronal-astrocytic co-culture system, where synthetic Aß peptides were added into the media without or with cotreatment with Aß12-28P, which is a nontoxic peptide antagonist of ApoE/Aß binding. Compared with neurons cultured alone, intraneuronal Aß content was significantly increased in neurons co-cultured with wild-type but not with ApoE knockout (KO) astrocytes. Neurons co-cultured with astrocytes also showed impaired intraneuronal degradation of Aß, increased level of intraneuronal Aß oligomers, and marked down-regulation of several synaptic proteins. Aß12-28P treatment significantly reduced intraneuronal Aß accumulation, including Aß oligomer level, and inhibited loss of synaptic proteins. Furthermore, we showed significantly reduced intraneuronal Aß accumulation in APPSW/PS1dE9/ApoE KO mice compared with APPSW/PS1dE9/ApoE targeted replacement mice that expressed various human ApoE isoforms. Data from our co-culture and in vivo experiments indicate an essential role of ApoE in the mechanism of intraneuronal Aß accumulation and provide evidence that ApoE/Aß binding antagonists can effectively prevent this process.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Synapses/pathology , Animals , Astrocytes/pathology , Cells, Cultured , Coculture Techniques , Endocytosis , Extracellular Space/metabolism , Female , Gene Knockdown Techniques , Hippocampus/pathology , Humans , Mice , Mice, Knockout , Nerve Degeneration/metabolism , Protein Binding , Protein Isoforms/metabolism , Subcellular Fractions/metabolism , Synapses/metabolism
12.
Neurosci Lett ; 538: 26-31, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23370284

ABSTRACT

Prionoses are a group of neurodegenerative diseases characterized by misfolding of cellular prion protein (PrP(C)) and accumulation of its diseases specific conformer PrP(Sc) in the brain and neuropathologically, they can be associated with presence or absence of PrP amyloid deposits. Functional molecular chaperones (MCs) that constitute the unfolded protein response include heat shock proteins and glucose-regulated protein families. They protect intracellular milieu against various stress conditions including accumulation of misfolded proteins and oxidative stress, typical of neurodegenerative diseases. Little is known about the role of MCs in pathogenesis of prionoses in mammalian prion model systems. In this study we characterized MCs response pattern in mice infected with various mouse adapted scrapie strains. Rather than uniform upregulation of MCs, we encountered two distinctly different patterns of MCs response distinguishing ME7 and 87V strains from 22L and 139A strains. ME7 and 87V strains are known for the induction of amyloid deposition in infected animals, while in mice infected with 22L and 139A strains amyloid deposits are absent. MCs response pattern similar to that associated with amyloidogenic ME7 and 87V strains was also observed in APPPS1-21 Alzheimer's transgenic mice, which represent an aggressive model of cerebral amyloidosis caused by ß-amyloid deposition. Our results highlight the probability that different mechanisms of MCs regulation exist driven by amyloidogenic and non-amyloidogenic nature of prion strains.


Subject(s)
Brain/metabolism , Molecular Chaperones/metabolism , Prions/physiology , Animals , Brain/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Female , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/pathology , Prions/pathogenicity
13.
Neurobiol Dis ; 34(2): 267-78, 2009 May.
Article in English | MEDLINE | ID: mdl-19385058

ABSTRACT

The pathogenesis of prion diseases is related to conformational transformation of cellular prion protein (PrP(C)) into a toxic, infectious, and self-replicating conformer termed PrP(Sc). Following extracerebral inoculation, the replication of PrP(Sc) is confined for months to years to the lymporeticular system (LRS) before the secondary CNS involvement results in occurrence of neurological symptoms. Therefore, replication of PrP(Sc), in the early stage of infection can be targeted by therapeutic approaches, which like passive immunization have limited blood-brain-barrier penetration. In this study, we show that 6D11 anti-PrP monoclonal antibody (Mab) prevents infection on a FDC-P1 myeloid precursor cell line stably infected with 22L mouse adapted scrapie strain. Passive immunization of extracerebrally infected CD-1 mice with Mab 6D11 resulted in effective suppression of PrP(Sc) replication in the LRS. Although, a rebound of PrP(Sc) presence occurred when the Mab 6D11 treatment was stopped, passively immunized mice showed a prolongation of the incubation period by 36.9% (pb0.0001) and a significant decrease in CNS pathology compared to control groups receiving vehicle or murine IgG. Our results indicate that antibody-based therapeutic strategies can be used, even on a short-term basis, to delay or prevent disease in subjects accidentally exposed to prions.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunization, Passive/methods , Lymphatic System/drug effects , Myeloid Progenitor Cells/drug effects , PrPSc Proteins/antagonists & inhibitors , Prion Diseases/drug therapy , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/immunology , Brain/metabolism , Cell Line , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Humans , Lymphatic System/immunology , Lymphatic System/metabolism , Mice , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/metabolism , PrPSc Proteins/biosynthesis , Prion Diseases/immunology , Prion Diseases/metabolism , Protein Conformation/drug effects , Scrapie/drug therapy , Scrapie/immunology , Scrapie/metabolism , Treatment Outcome
14.
Proc Natl Acad Sci U S A ; 103(49): 18787-92, 2006 Dec 05.
Article in English | MEDLINE | ID: mdl-17116874

ABSTRACT

The amyloid-beta (Abeta) cascade hypothesis of Alzheimer's disease (AD) maintains that accumulation of Abeta peptide constitutes a critical event in the early disease pathogenesis. The direct binding between Abeta and apolipoprotein E (apoE) is an important factor implicated in both Abeta clearance and its deposition in the brain's parenchyma and the walls of meningoencephalic vessels as cerebral amyloid angiopathy. With the aim of testing the effect of blocking the apoE/Abeta interaction in vivo as a potential novel therapeutic target for AD pharmacotherapy, we have developed Abeta12-28P, which is a blood-brain-barrier-permeable nontoxic, and nonfibrillogenic synthetic peptide homologous to the apoE binding site on the full-length Abeta. Abeta12-28P binds with high affinity to apoE, preventing its binding to Abeta, but has no direct effect on Abeta aggregation. Abeta12-28P shows a strong pharmacological effect in vivo. Its systemic administration resulted in a significant reduction of Abeta plaques and cerebral amyloid angiopathy burden and a reduction of the total brain level of Abeta in two AD transgenic mice models. The treatment did not affect the levels of soluble Abeta fraction or Abeta oligomers, indicating that inhibition of the apoE/Abeta interaction in vivo has a net effect of increasing Abeta clearance over deposition and at the same time does not create conditions favoring formation of toxic oligomers. Furthermore, behavioral studies demonstrated that treatment with Abeta12-28P prevents a memory deficit in transgenic animals. These findings provide evidence of another therapeutic approach for AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Apolipoproteins E/antagonists & inhibitors , Apolipoproteins E/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Binding, Competitive/physiology , Female , Mice , Mice, Transgenic , Peptide Fragments/metabolism
15.
Eur J Neurosci ; 23(10): 2635-47, 2006 May.
Article in English | MEDLINE | ID: mdl-16817866

ABSTRACT

Prion diseases are transmissible and invariably fatal neurodegenerative disorders associated with a conformational transformation of the cellular prion protein (PrP(C)) into a self-replicating and proteinase K (PK)-resistant conformer, scrapie PrP (PrP(Sc)). Humoral immunity may significantly prolong the incubation period and even prevent disease in murine models of prionoses. However, the mechanism(s) of action of anti-PrP monoclonal antibodies (Mabs) remain(s) obscure. The murine neuroblastoma N2a cell line, infected with the 22L mouse-adapted scrapie strain, was used to screen a large library of Mabs with similar binding affinities to PrP, to identify those antibodies which could clear established infection and/or prevent infection de novo. Three Mabs were found capable of complete and persistent clearing of already-infected N2a cells of PrP(Sc). These antibodies were 6D11 (generated to PK-resistant PrP(Sc) and detecting PrP residues 93-109), and 7H6 and 7A12, which were raised against recombinant PrP and react with neighbouring epitopes of PrP residues 130-140 and 143-155, respectively. Mabs were found to interact with PrP(Sc) formation both on the cell surface and after internalization in the cytosol. Treatment with Mabs was not associated with toxicity nor did it result in decreased expression of PrP(C). Both preincubation of N2a cells with Mabs prior to exposure to 22L inoculum and preincubation of the inoculum with Mabs prior to infecting N2a cells resulted in a significant reduction in PrP(Sc) levels. Information provided in these studies is important for the rational design of humoral immune therapy for prion infection in animals and eventually in humans.


Subject(s)
Antibodies, Monoclonal/pharmacology , PrPC Proteins/drug effects , PrPSc Proteins/drug effects , Prion Diseases/immunology , Prion Diseases/prevention & control , Animals , Antibody Affinity , Cells, Cultured , Epitope Mapping , Immunodominant Epitopes , Mice , PrPC Proteins/immunology , PrPSc Proteins/immunology , PrPSc Proteins/pathogenicity
16.
Am J Pathol ; 165(3): 937-48, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331417

ABSTRACT

Alzheimer's disease (AD) is associated with accumulation of beta-amyloid (Abeta). A major genetic risk factor for sporadic AD is inheritance of the apolipoprotein (apo) E4 allele. ApoE can act as a pathological chaperone of Abeta, promoting its conformational transformation from soluble Abeta into toxic aggregates. We determined if blocking the apoE/Abeta interaction reduces Abeta load in transgenic (Tg) AD mice. The binding site of apoE on Abeta corresponds to residues 12 to 28. To block binding, we synthesized a peptide containing these residues, but substituted valine at position 18 to proline (Abeta12-28P). This changed the peptide's properties, making it non-fibrillogenic and non-toxic. Abeta12-28P competitively blocks binding of full-length Abeta to apoE (IC50 = 36.7 nmol). Furthermore, Abeta12-28P reduces Abeta fibrillogenesis in the presence of apoE, and Abeta/apoE toxicity in cell culture. Abeta12-28P is blood-brain barrier-permeable and in AD Tg mice inhibits Abeta deposition. Tg mice treated with Abeta12-28P for 1 month had a 63.3% reduction in Abeta load in the cortex (P = 0.0043) and a 59.5% (P = 0.0087) reduction in the hippocampus comparing to age-matched control Tg mice. Antibodies against Abeta were not detected in sera of treated mice; therefore the observed therapeutic effect of Abeta12-28P cannot be attributed to an antibody clearance response. Our experiments demonstrate that compounds blocking the interaction between Abeta and its pathological chaperones may be beneficial for treatment of beta-amyloid deposition in AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Peptide Fragments/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Apolipoproteins E/genetics , Astrocytes/metabolism , Astrocytes/pathology , Binding Sites/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Competitive Bidding , Female , Half-Life , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Neuroblastoma/metabolism , Neuroblastoma/pathology , Presenilin-1 , Protein Binding/drug effects , Tumor Cells, Cultured
17.
J Neuropathol Exp Neurol ; 63(7): 775-84, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15290902

ABSTRACT

The diagnosis of prion diseases in humans is challenging due to a lack of specific and sensitive non-invasive tests. Many forms of human prion disease including variant Creutzfeldt-Jakob disease (vCJD), Gerstmann-Sträussler-Scheinker (GSS) syndrome, and 10% of sporadic CJD cases are associated with amyloid deposition. Several positron emission tomography (PET) ligands have recently been developed to directly image beta-amyloid associated with Alzheimer disease. One of them, methoxy-X04, is a fluorescent derivative of Congo red with high binding affinity toward amyloid fibrils and good blood-brain barrier permeability. Using methoxy-X04, we investigated whether amyloid-targeting ligands can be also employed for direct imaging of amyloid deposits associated with some prion diseases. Such a method could potentially become a novel diagnostic approach for these conditions. Studies were performed on MB mice infected with the 87V mouse-adapted scrapie strain. Labeling of PrP amyloid plaques in brains of presymptomatic and symptomatic mice was demonstrated using in vivo transcranial two-photon microscopy after systemic administration of methoxy-X04. During real-time imaging, PrP amyloid deposits could be clearly distinguished 15 min after intravenous administration of methoxy-X04. The ligand showed rapid clearance from brain areas that did not contain amyloid deposits. PrP amyloid deposits could also be detected by direct application of methoxy-X04 on cerebellar sections from GSS patients. These results suggest that methoxy-X04 or similar derivatives could be used as PET imaging agents to improve the diagnosis of human prion diseases associated with amyloid deposition.


Subject(s)
Alkenes , Amyloid beta-Peptides/analysis , Benzene Derivatives , Plaque, Amyloid/pathology , Prion Diseases/diagnostic imaging , Prion Diseases/pathology , Tomography, Emission-Computed/methods , Alkenes/metabolism , Alkenes/pharmacokinetics , Animals , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacokinetics , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Mice , Plaque, Amyloid/metabolism , Predictive Value of Tests , Prion Diseases/metabolism , Reproducibility of Results , Scrapie/diagnostic imaging , Scrapie/metabolism , Scrapie/pathology , Stilbenes
18.
Neurochem Res ; 29(6): 1257-66, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15176482

ABSTRACT

The major neuropathological lesions defining Alzheimer's disease (AD) include neurofibrillary tangles and amyloid plaques, which are mainly composed of abnormally phosphorylated tau and amyloid-beta (A beta), respectively. Numerous neuropathological and neuroimaging studies indicate that at least one-third of AD cases are complicated by some degree of vascular pathology, whereas in a similar proportion of patients clinically diagnosed with vascular dementia, AD pathology is also present. Many classical vascular risk factors such as hypertension, diabetes mellitus, and hypercholesterolemia have recently been shown also to increase the risk of AD. Growing evidence suggests that vascular pathology lowers the threshold for the clinical presentation of dementia at a given level of AD-related pathology and potentially directly promotes AD lesions such as A beta plaques. Cerebral ischemia, chronically up-regulates expression of the amyloid precursor protein (APP), which is the precursor to the amyloid beta peptide and damages the blood-brain barrier (BBB), affecting A beta peptide clearance from the brain. Recognition of the importance of these vascular risk factors for AD-related dementia and their treatment will be beneficial not only for preventing cardiac, cerebral, and peripheral complications of vascular disease, but also will likely have a direct impact on the occurrence of sporadic AD in older subjects. In this paper, we review some of the links between vascular risk factors and AD pathology and present data on the direct effect of ischemia on cognitive function and A beta deposition in a mouse model of AD.


Subject(s)
Alzheimer Disease/pathology , Dementia, Vascular/pathology , Amyloid/metabolism , Animals , Brain/pathology , Crosses, Genetic , Dementia, Vascular/psychology , Disease Models, Animal , Humans , Maze Learning , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurons/pathology , Risk Factors , Schizophrenic Psychology
19.
J Neuropathol Exp Neurol ; 63(5): 418-28, 2004 May.
Article in English | MEDLINE | ID: mdl-15198121

ABSTRACT

In Alzheimer disease (AD) patients, early memory dysfunction is associated with glucose hypometabolism and neuronal loss in the hippocampus. Double transgenic (Tg) mice co-expressing the M146L presenilin 1 (PS1) and K670N/M671L, the double "Swedish" amyloid precursor protein (APP) mutations, are a model of AD amyloid-beta deposition (Abeta) that exhibits earlier and more profound impairments of working memory and learning than single APP mutant mice. In this study we compared performance on spatial memory tests, regional glucose metabolism, Abeta deposition, and neuronal loss in APP/PS1, PS1, and non-Tg (nTg) mice. At the age of 2 months no significant morphological and metabolic differences were detected between 3 studied genotypes. By 8 months, however, APP/PS1 mice developed selective impairment of spatial memory, which was significantly worse at 22 months and was accompanied by reduced glucose utilization in the hippocampus and a 35.8% dropout of neurons in the CA1 region. PS1 mice exhibited a similar degree of neuronal loss in CA1 but minimal memory deficit and no impairment of glucose utilization compared to nTg mice. Deficits in 22 month APP/PS1 mice were accompanied by a substantially elevated Abeta load, which rose from 2.5% +/- 0.4% at 8 months to 17.4% +/- 4.6%. These findings implicate Abeta or APP in the behavioral and metabolic impairments in APP/PS1 mice and the failure to compensate functionally for PS1-related hippocampal cell loss.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Glucose/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Memory Disorders/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Count , Cell Death/genetics , Disease Models, Animal , Down-Regulation/genetics , Female , Hippocampus/pathology , Hippocampus/physiopathology , Male , Maze Learning/physiology , Membrane Proteins/genetics , Memory Disorders/genetics , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Motor Activity/genetics , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/pathology , Presenilin-1
20.
Dement Geriatr Cogn Disord ; 16(4): 283-6, 2003.
Article in English | MEDLINE | ID: mdl-14512725

ABSTRACT

Inflammatory mechanisms are involved in the pathogenesis of Alzheimer's disease (AD). It is postulated that cytokine synthesis is altered in AD patients compared with nondemented subjects. Glucocorticoids play an important role in cytokine synthesis. We assessed the release of tumor necrosis factor-alpha (TNF-alpha), interleukin-10 (IL-10) and interleukin-12 (IL-12) and its regulation by dexamethasone in AD patients in vitro. Cytokine levels were measured using the ELISA method in unstimulated, LPS-stimulated or whole-blood samples incubated with LPS and dexamethasone from 18 AD patients and 12 controls. The cytokine levels spontaneously produced by blood cells after incubation with LPS or LPS and dexamethasone did not differ significantly between groups. Dexamathasone inhibited TNF-alpha synthesis by LPS-stimulated blood cells more effectively in AD patients than in controls. These results suggest that cytokine synthesis in AD patients could be regulated by glucocorticoids in a different way than in nondemented subjects.


Subject(s)
Alzheimer Disease/metabolism , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Humans , In Vitro Techniques , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...