Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Acta Neuropathol ; 144(1): 143-153, 2022 07.
Article in English | MEDLINE | ID: mdl-35581440

ABSTRACT

Amyloid beta (Aß) plaque is a defining pathologic feature of Alzheimer disease (AD). Aducanumab, a monoclonal IgG1 that selectively binds aggregated species of Aß, has been shown by amyloid positron emission tomography (Amyloid PET) to reduce Aß plaques in patients with prodromal and mild AD. This is the first autopsy report of the AD neuropathology in a patient previously treated with aducanumab. The patient was an 84-year-old woman who was randomized to the placebo arm of the PRIME Phase 1b study (221AD103). The patient progressed to moderate dementia (MMSE = 14/30), beyond the targeted early AD treatment stage, before receiving aducanumab in the long-term extension (LTE). The patient then received 32 monthly doses of aducanumab, titrated up to 6 mg/kg, for a cumulative dose of 186 mg/kg. In the LTE, Amyloid PET scans demonstrated robust Aß plaque reduction, from a composite standard uptake value ratio (SUVR) of 1.5 at screening to < 1.1 at 56 weeks post-aducanumab dosing. MRI examinations were negative for amyloid-related imaging abnormalities (ARIA). She passed away in hospice care 4 months after her last dose of aducanumab. The postmortem neuropathologic examination confirmed AD neuropathologic changes. Aß and IBA1 immunohistochemistry assays demonstrated sparse residual Aß plaque engaged by amoeboid reactive microglia. Phospho-Tau (pTau) immunohistochemistry demonstrated neocortical neurofibrillary degeneration (Braak stage V, NIA/AA Stage B3). However, the density of pTau neuropathology, including neuritic plaque pTau (NP-Tau), appeared lower in the PRIME LTE Patient compared to a reference cohort of untreated Braak stage V-VI, NIA/AA Stage B3 AD cases. Taken together, this case report is the first to provide Amyloid PET and neuropathologic evidence substantiating the impact of aducanumab to reduce Aß plaque neuropathology in a patient with AD. Furthermore, this report underscores the critical importance of autopsy neuropathology studies to augment our understanding of aducanumab's mechanism of action and impact on AD biomarkers.


Subject(s)
Alzheimer Disease , Antibodies, Monoclonal, Humanized , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Clinical Trials, Phase I as Topic , Female , Humans , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Plaque, Amyloid/prevention & control , Positron-Emission Tomography , Randomized Controlled Trials as Topic
2.
CPT Pharmacometrics Syst Pharmacol ; 11(3): 362-372, 2022 03.
Article in English | MEDLINE | ID: mdl-35029320

ABSTRACT

Alzheimer's disease (AD) is an irreversible, progressive brain disorder that impairs memory and cognitive function. Dysregulation of the amyloid-ß (Aß) pathway and amyloid plaque accumulation in the brain are hallmarks of AD. Aducanumab is a human, immunoglobulin gamma 1 monoclonal antibody targeting aggregated forms of Aß. In phase Ib and phase III studies, aducanumab reduced Aß plaques in a dose dependent manner, as measured by standard uptake value ratio of amyloid positron emission tomography imaging. The goal of this work was to develop a quantitative systems pharmacology model describing the production, aggregation, clearance, and transport of Aß as well as the mechanism of action for the drug to understand the relationship between aducanumab dosing regimens and changes of different Aß species, particularly plaques in the brain. The model was used to better understand the pharmacodynamic effects observed in the clinical trials of aducanumab and assist in the clinical development of future Aß therapies.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized , Brain/metabolism , Humans , Network Pharmacology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism
3.
Nat Neurosci ; 23(12): 1580-1588, 2020 12.
Article in English | MEDLINE | ID: mdl-33199898

ABSTRACT

Amyloid-ß (Aß) deposits are a relatively late consequence of Aß aggregation in Alzheimer's disease. When pathogenic Aß seeds begin to form, propagate and spread is not known, nor are they biochemically defined. We tested various antibodies for their ability to neutralize Aß seeds before Aß deposition becomes detectable in Aß precursor protein-transgenic mice. We also characterized the different antibody recognition profiles using immunoprecipitation of size-fractionated, native, mouse and human brain-derived Aß assemblies. At least one antibody, aducanumab, after acute administration at the pre-amyloid stage, led to a significant reduction of Aß deposition and downstream pathologies 6 months later. This demonstrates that therapeutically targetable pathogenic Aß seeds already exist during the lag phase of protein aggregation in the brain. Thus, the preclinical phase of Alzheimer's disease-currently defined as Aß deposition without clinical symptoms-may be a relatively late manifestation of a much earlier pathogenic seed formation and propagation that currently escapes detection in vivo.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Brain Chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurofilament Proteins/cerebrospinal fluid , Plaque, Amyloid/pathology , Tissue Extracts/pharmacology
4.
Nat Struct Mol Biol ; 27(12): 1125-1133, 2020 12.
Article in English | MEDLINE | ID: mdl-32989305

ABSTRACT

The amyloid cascade hypothesis, according to which the self-assembly of amyloid-ß peptide (Aß) is a causative process in Alzheimer's disease, has driven many therapeutic efforts for the past 20 years. Failures of clinical trials investigating Aß-targeted therapies have been interpreted as evidence against this hypothesis, irrespective of the characteristics and mechanisms of action of the therapeutic agents, which are highly challenging to assess. Here, we combine kinetic analyses with quantitative binding measurements to address the mechanism of action of four clinical stage anti-Aß antibodies, aducanumab, gantenerumab, bapineuzumab and solanezumab. We quantify the influence of these antibodies on the aggregation kinetics and on the production of oligomeric aggregates and link these effects to the affinity and stoichiometry of each antibody for monomeric and fibrillar forms of Aß. Our results reveal that, uniquely among these four antibodies, aducanumab dramatically reduces the flux of Aß oligomers.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Humans , Kinetics , Models, Biological , Models, Molecular , Neuroprotective Agents/chemistry , Peptide Fragments/chemistry , Peptide Mapping/methods , Protein Aggregates/drug effects , Protein Conformation , Structure-Activity Relationship
5.
Sci Rep ; 8(1): 6412, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686315

ABSTRACT

Aducanumab, a human-derived antibody targeting amyloid-ß (Aß), is in Phase 3 clinical trials for the treatment of Alzheimer's disease. Biochemical and structural analyses show that aducanumab binds a linear epitope formed by amino acids 3-7 of the Aß peptide. Aducanumab discriminates between monomers and oligomeric or fibrillar aggregates based on weak monovalent affinity, fast binding kinetics and strong avidity for epitope-rich aggregates. Direct comparative studies with analogs of gantenerumab, bapineuzumab and solanezumab demonstrate clear differentiation in the binding properties of these antibodies. The crystal structure of the Fab fragment of aducanumab bound to its epitope peptide reveals that aducanumab binds to the N terminus of Aß in an extended conformation, distinct from those seen in structures with other antibodies that target this immunodominant epitope. Aducanumab recognizes a compact epitope that sits in a shallow pocket on the antibody surface. In silico analyses suggest that aducanumab interacts weakly with the Aß monomer and may accommodate a variety of peptide conformations, further supporting its selectivity for Aß aggregates. Our studies provide a structural rationale for the low affinity of aducanumab for non-pathogenic monomers and its greater selectivity for aggregated forms than is seen for other Aß-targeting antibodies.


Subject(s)
Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Binding Sites, Antibody , Enzyme-Linked Immunosorbent Assay , Humans , Immunotherapy , Kinetics , Molecular Docking Simulation , Protein Conformation , Surface Plasmon Resonance
7.
Am J Pathol ; 187(6): 1399-1412, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28408124

ABSTRACT

The clinical progression of Alzheimer disease (AD) is associated with the accumulation of tau neurofibrillary tangles, which may spread throughout the cortex by interneuronal tau transfer. If so, targeting extracellular tau species may slow the spreading of tau pathology and possibly cognitive decline. To identify suitable target epitopes, we tested the effects of a panel of tau antibodies on neuronal uptake and aggregation in vitro. Immunodepletion was performed on brain extract from tau-transgenic mice and postmortem AD brain and added to a sensitive fluorescence resonance energy transfer-based tau uptake assay to assess blocking efficacy. The antibodies reduced tau uptake in an epitope-dependent manner: N-terminal (Tau13) and middomain (6C5 and HT7) antibodies successfully prevented uptake of tau species, whereas the distal C-terminal-specific antibody (Tau46) had little effect. Phosphorylation-dependent (40E8 and p396) and C-terminal half (4E4) tau antibodies also reduced tau uptake despite removing less total tau by immunodepletion, suggesting specific interactions with species involved in uptake. Among the seven antibodies evaluated, 6C5 most efficiently blocked uptake and subsequent aggregation. More important, 6C5 also blocked neuron-to-neuron spreading of tau in a unique three-chamber microfluidic device. Furthermore, 6C5 slowed down the progression of tau aggregation even after uptake had begun. Our results imply that not all antibodies/epitopes are equally robust in terms of blocking tau uptake of human AD-derived tau species.


Subject(s)
Alzheimer Disease/metabolism , Neurons/metabolism , tau Proteins/metabolism , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Brain/metabolism , Brain/pathology , Cells, Cultured , Epitopes/immunology , Female , Humans , Interneurons/metabolism , Male , Mice, Transgenic , Microfluidic Analytical Techniques , Molecular Targeted Therapy/methods , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation , tau Proteins/antagonists & inhibitors , tau Proteins/immunology
8.
J Neurosci ; 36(50): 12549-12558, 2016 12 14.
Article in English | MEDLINE | ID: mdl-27810931

ABSTRACT

Calcium homeostasis plays a major role in maintaining neuronal function under physiological conditions. Amyloid-ß (Aß) initiates pathological processes that include disruption in intracellular calcium levels, so amelioration of the calcium alteration could serve as an indirect functional indicator of treatment efficacy. Therefore, calcium dynamics were used as a measure of functional outcome. We evaluated the effects of the anti-Aß antibody aducanumab on calcium homeostasis and plaque clearance in aged Tg2576 mice with in vivo multiphoton imaging. Acute topical application of aducanumab to the brain resulted in clearance of amyloid plaques. Although chronic systemic administration of aducanumab in 22-month-old mice did not clear existing plaques, calcium overload was ameliorated over time. Therefore, this antibody likely restores neuronal network function that possibly underlies cognitive deficits, indicating promise as a clinical treatment. In addition, functional readouts such as calcium overload may be a more useful outcome measure to monitor treatment efficacy in models of Alzheimer's disease compared with amyloid burden alone. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is currently without a cure. Aducanumab is an anti-amyloid-ß antibody being developed for the treatment of AD. Interim analyses of a phase 1b clinical trial have suggested potential beneficial effects on amyloid pathology and cognitive status in patients treated with aducanumab (Sevigny et al., 2016). Here, we show that a murine analog of aducanumab clears amyloid plaques in an acute setting and restores calcium homeostasis disrupted in a mouse model of AD upon chronic treatment. Therefore, we demonstrate that aducanumab reverses a functional outcome measure reflective of neural network activity.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Calcium/metabolism , Homeostasis/drug effects , Immunotherapy/methods , Aging/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Down-Regulation , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Nerve Net/drug effects , Nerve Net/physiopathology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Receptors, N-Methyl-D-Aspartate/biosynthesis
9.
Nature ; 537(7618): 50-6, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27582220

ABSTRACT

Alzheimer's disease (AD) is characterized by deposition of amyloid-ß (Aß) plaques and neurofibrillary tangles in the brain, accompanied by synaptic dysfunction and neurodegeneration. Antibody-based immunotherapy against Aß to trigger its clearance or mitigate its neurotoxicity has so far been unsuccessful. Here we report the generation of aducanumab, a human monoclonal antibody that selectively targets aggregated Aß. In a transgenic mouse model of AD, aducanumab is shown to enter the brain, bind parenchymal Aß, and reduce soluble and insoluble Aß in a dose-dependent manner. In patients with prodromal or mild AD, one year of monthly intravenous infusions of aducanumab reduces brain Aß in a dose- and time-dependent manner. This is accompanied by a slowing of clinical decline measured by Clinical Dementia Rating-Sum of Boxes and Mini Mental State Examination scores. The main safety and tolerability findings are amyloid-related imaging abnormalities. These results justify further development of aducanumab for the treatment of AD. Should the slowing of clinical decline be confirmed in ongoing phase 3 clinical trials, it would provide compelling support for the amyloid hypothesis.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Plaque, Amyloid/drug therapy , Plaque, Amyloid/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Brain/drug effects , Brain/metabolism , Clinical Trials, Phase III as Topic , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Models, Biological , Plaque, Amyloid/pathology , Protein Aggregation, Pathological/drug therapy , Solubility
10.
Neuropharmacology ; 103: 57-68, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26690893

ABSTRACT

Reducing the production of larger aggregation-prone amyloid ß-peptides (Aß) remains an untested therapeutic approach for reducing the appearance and growth of Aß plaques in the brain, which are a hallmark pathological feature of Alzheimer's disease. γ-Secretase modulators (GSMs) are therapeutics that impact γ-secretase-dependent cleavage of amyloid precursor protein to promote the production of shorter Aß peptides that are less prone to aggregation and plaque deposition. This is accomplished without inhibiting overall γ-secretase function and cleavage of other substrates, which is believed to be a source of deleterious side effects. Here, we report the pharmacokinetic and pharmacodynamic properties of BIIB042, a novel bioavailable and brain-penetrant GSM. In cell-based assays, BIIB042 reduced the levels of Aß42, increased the levels of Aß38 and had little effect on the levels of Aß40, the most abundant Aß species. Similar pharmacodynamic properties were confirmed in the central nervous system and in plasma of mice and rats, and also in plasma of cynomolgus monkeys after a single oral dose of BIIB042. BIIB042 reduced Aß42 levels and Aß plaque burden in Tg2576 mice, which overexpress human amyloid precursor protein and serve as a model system for Alzheimer's disease. BIIB042 did not inhibit cleavage of other γ-secretase substrates in cell-based and in vivo signaling and cleavage assays. The pharmacodynamic effects of lowering Aß42 in the central nervous system coupled with demonstrated efficacy in reducing plaque pathology suggests modulation of γ-secretase, with molecules like BIIB042, is a compelling therapeutic approach for the treatment of Alzheimer's disease.


Subject(s)
Aldehydes/pharmacokinetics , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/enzymology , Aldehydes/administration & dosage , Amyloid beta-Peptides/blood , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Macaca fascicularis , Male , Mice , Plaque, Amyloid/metabolism , Protein Isoforms/blood , Rats , Rats, Inbred F344
11.
Neurobiol Aging ; 27(2): 270-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16399212

ABSTRACT

The relative contribution of Alzheimer's disease (AD) hippocampal neuronal pathology in cognitive decline is still a matter of debate. To address this issue, we performed a stereological analysis of layer II of the entorhinal cortex and the CA1 field of the hippocampus in 34 autopsy cases covering the whole spectrum of old age and Clinical Dementia Rating (CDR) scores. In both areas, the proportion of neurofibrillary tangle (NFT)-containing neurons increased steadily as a function of the CDR score. Questionable dementia was associated with a 1.9% neuronal loss in the entorhinal cortex and 26% in the CA1 field. NFT numbers predicted only 38% of the neuron number variability in the entorhinal cortex and 55% in the CA1 field. Neuron counts in the entorhinal cortex and both neuron and NFT counts in the CA1 field were significantly associated with cognitive status explaining 25% and 44% of the CDR variability, respectively. Our data reveal a dissociation between the patterns of progression of NFT and neuronal loss in the entorhinal cortex and CA1 field. Moreover, they show that less than 50% of the cognitive variability may be attributable to AD neuronal pathology in these areas.


Subject(s)
Alzheimer Disease/pathology , Cognition Disorders/pathology , Entorhinal Cortex/pathology , Hippocampus/pathology , Neurons/pathology , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/complications , Brain Mapping , Cell Count/methods , Cognition Disorders/etiology , Diagnosis, Computer-Assisted/methods , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Neurofibrillary Tangles/pathology
12.
Am J Pathol ; 165(3): 987-95, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331422

ABSTRACT

Transgenic mice mimicking certain features of Alzheimer's disease (AD)-pathology, namely amyloid plaques and neurofibrillary tangles, have been developed in an effort to better understand the mechanism leading to the formation of these characteristic cerebral lesions. More recently, these animal models have been widely used to investigate emergent therapies aimed at the reduction of the cerebral amyloid load. Several studies have shown that immunotherapy targeting the amyloid peptide (Abeta) is efficacious at clearing the amyloid plaques or preventing their formation, and at reducing the memory/behavior impairment observed in these animals. In AD, different types of plaques likely have different pathogenic significance, and further characterization of plaque pathology in the PDAPP transgenic mice would enhance the evaluation of potential therapeutics. In the present study, a morphological classification of amyloid plaques present in the brains of PDAPP mice was established by using Thioflavin-S staining. Neuritic dystrophy associated with amyloid plaques was also investigated. Finally, the efficacy of passive immunization with anti-Abeta antibodies on the clearance of Thio-S positive amyloid plaques was studied. Our results show that distinct morphological types of plaques are differentially cleared depending upon the isotype of the antibody.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/therapeutic use , Immunotherapy , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Thiazoles/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Benzothiazoles , Brain/pathology , Disease Models, Animal , Fluorescent Dyes , Heterozygote , Humans , Immunization, Passive , Metabolic Clearance Rate , Mice , Mice, Transgenic , Neurites/metabolism , Neurites/pathology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology
13.
J Comp Neurol ; 463(3): 281-302, 2003 Aug 25.
Article in English | MEDLINE | ID: mdl-12820162

ABSTRACT

We performed a stereologic analysis of a subset of pyramidal neurons known to be vulnerable in Alzheimer's disease (AD) and characterized by particularly high somatodendritic levels of nonphosphorylated neurofilament protein. In the neocortex, these large pyramidal neurons reside in the deep part of layer III (layer IIIc) and the superficial part of layer V (layer Va). We focused on prefrontal cortex area 9 in elderly control cases in comparison to cases with different degrees of cognitive dysfunction. The results confirmed that these neurons are preferentially vulnerable in AD, as their numbers decrease dramatically in cases with definite dementia, correlating strongly with the severity of the disease, to a nearly complete loss (>90%) in the endstages of AD. Furthermore, a triple-labeling experimental paradigm revealed that these particular neurons are far more likely to develop neurofibrillary tangles (NFT) and do so at a faster rate than other pyramidal cells. Nonphosphorylated neurofilament protein-rich neurons also shrink considerably during formation of NFT and the largest among them are preferentially affected. Laminar differences in the severity of these effects were observed, layer Va being more severely affected, possibly correlating with the involvement of specific cortical projections. These data reveal that different populations of neurons prone to NFT formation are lost at different rates in AD, and that nonphosphorylated neurofilament protein-enriched neurons emerge as a strikingly vulnerable subpopulation of neurons. Their preferential involvement suggests that neurons providing specific corticocortical connections between association areas are at high risk for degeneration in AD.


Subject(s)
Alzheimer Disease/pathology , Nerve Degeneration/pathology , Prefrontal Cortex/pathology , Pyramidal Cells/pathology , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Cell Count , Cognition Disorders/physiopathology , Female , Humans , Immunohistochemistry , Male , Neurofibrillary Tangles/pathology , Neurofilament Proteins/metabolism , Prognosis , Pyramidal Cells/metabolism , Serum Amyloid P-Component/metabolism
14.
Biochim Biophys Acta ; 1619(2): 167-76, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12527113

ABSTRACT

Both phosphorylation and O-GlcNAc glycosylation posttranslationally modify microtubule-associated Tau proteins. Whereas the hyperphosphorylation of these proteins that occurs in Alzheimer's disease is well characterized, little is known about the O-GlcNAc glycosylation. The present study demonstrates that a balance exists between phosphorylation and O-GlcNAc glycosylation of Tau proteins, and furthermore that a dysfunction of this balance correlates with reduced nuclear localization. The affinity of Tau proteins for WGA lectin, together with evidence from [3H]-galactose transfer and analysis of beta-eliminated products, demonstrated the presence of O-GlcNAc residues on both cytosolic and nuclear Tau proteins. In addition, our data indicated the existence of a balance between phosphorylation and O-GlcNAc glycosylation events. Indeed, as demonstrated by 2D-electrophoresis and Western blotting, O-GlcNAc residues were mainly located on the less phosphorylated Tau 441 variants, whereas the more phosphorylated forms were devoid of O-GlcNAc residues. Furthermore, the Tau protein hyperphosphorylation induced by cellular okadaic acid treatment was correlated with reduced incorporation of O-GlcNAc residues into Tau proteins and with diminished Tau transfer into the nucleus. Hence, this paper establishes a direct relationship between O-GlcNAc glycosylation, phosphorylation and cellular localization of Tau proteins.


Subject(s)
Cell Nucleus/metabolism , tau Proteins/metabolism , Blotting, Western , Cytosol/metabolism , Electrophoresis, Gel, Two-Dimensional , Enzyme Inhibitors/pharmacology , Glycosylation/drug effects , Humans , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/metabolism , Okadaic Acid/pharmacology , Phosphorylation , Protein Isoforms/metabolism , Tumor Cells, Cultured , tau Proteins/biosynthesis , tau Proteins/genetics
15.
J Neuropathol Exp Neurol ; 62(1): 55-67, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12528818

ABSTRACT

The entorhinal cortex and hippocampus are the first cortical regions to be affected by the degenerative cellular process that leads to Alzheimer disease (AD) and display a limited degree of neuronal alterations in normal aging. Several quantitative studies have reported a substantial loss of neurons in these regions and a parallel increase in the number of neurofibrillary tangles (NFTs). However, accurate quantitative data on the dynamics of NFT formation are lacking. Here, we performed a stereologic assessment of the proportions of intracellular and extracellular (ghost) NFTs (iNFTs and eNFTs, respectively) and unaffected neurons in layer II of the entorhinal cortex and in the pyramidal cell layer of the CA1 field of the hippocampus in elderly control cases compared to cases with varying degrees of cognitive dysfunction. The data revealed differential rates of formation of iNFTs and eNFTs between the 2 regions and confirmed the presence of a severe disease-associated, but not age-related, neuronal loss. They also revealed that large numbers of neurons may persist either unaffected or in a transitional stage of NFT formation until the late stages of AD progression. These neurons with viability potential constitute 73% of the total numbers of profiles in layer II of the entorhinal cortex and 77% in the CA1 field in cases with a Clinical Dementia Rating score of 3. Whereas it is not possible in the present study to assess how functional such neurons with altered physiology might be, it is nonetheless likely that these transitional neurons open new options for potential therapeutic interventions aimed at protecting neurons vulnerable to neurofibrillary degeneration.


Subject(s)
Alzheimer Disease/pathology , Entorhinal Cortex/pathology , Neurons/pathology , Neurons/physiology , Aged , Aged, 80 and over , Cell Count/methods , Cell Survival/physiology , Disease Progression , Entorhinal Cortex/chemistry , Female , Humans , Male , Middle Aged , Neurofibrillary Tangles/pathology , Neurons/chemistry , Stereotaxic Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...