Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
2.
Neurosci Behav Physiol ; 36(8): 789-99, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964455

ABSTRACT

The processes of learning and storage of the results of learning were studied in a model of Alzheimer's disease in two groups of rhesus macaques (three individuals in each group). Studies were performed after injection of neurotoxins (group I) and physiological saline (group II, controls). Two months after injections (stage C1), learning parameters were studied in monkeys of both groups using a new stimulus discrimination test (filled geometrical figures versus outline figures). There were significant differences between the animals of the two groups. Learning was hindered in monkeys of group I, with significant increases in the learning time (the time to achieve a stable probability of correct responding of 0.85) and in the probability of refusals. Monkeys of group II showed no learning impairment. Animals were trained to discriminate new stimuli (images of two monkeys) six months after injections (stage C3). Learning was impaired in animals of group I, such that learning measures had the same levels as previously; monkeys of group II showed no learning impairment. Analysis of the characteristics of working memory, which is involved in storing the results of new learning, was performed at stage C1; monkeys of group I showed significant degradation of these measures, with a significant decrease in the probability of correct solutions at stage C1 (to a level of 0.5), with some increase at stages C2 (at four months) and C3, along with a significant increase in the probability of refusals, values being similar at all time points. For monkeys of group II, these characteristics showed no degradation. Motor response times at stages C1, C2, and C3 were not different for the two groups of monkeys. The structural-functional organization of interactions between sensory and cognitive processes during learning and the storage of information in working memory are discussed, as is the role of the associative areas of the cortex in these interactions.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Learning Disabilities/etiology , Memory, Short-Term/physiology , Alzheimer Disease/chemically induced , Animals , Behavior, Animal , Discrimination Learning/physiology , Disease Models, Animal , Macaca mulatta , Neurotoxins , Pattern Recognition, Visual/drug effects , Pattern Recognition, Visual/physiology , Photic Stimulation/methods , Probability , Reaction Time/drug effects , Reaction Time/physiology
3.
Ross Fiziol Zh Im I M Sechenova ; 91(8): 857-71, 2005 Aug.
Article in Russian | MEDLINE | ID: mdl-16252682

ABSTRACT

Processes of novelty learning and keeping the results in Alzheimer's disease in two groups of rhesus-monkeys (three monkeys in each group), were studied: following neurotoxins administrati- on (I group) and saline administration (II group). In two months after the injections (the C1 stage), considerable differences between the groups were revealed in the task of differentiation among contour shapes. For the I group monkeys the learning was difficult: the correct decision making did not reach 85 %, and the probability of refusing to make a decision increased. For the II group monkeys the learning characteristics were not disturbed. In six months after the injections (the C3 stage) the differences between the groups in the task of differentiation among new stimuli (heads of two monkeys) remained at the same level. When studying characteristics of the operative memory associated with keeping the learning results achieved at the C1 stage, a considerable worsening of these characteristics was revealed: diminishing of the correct decision making probability at the C1 stage (actually to the level of 0.5), increase in the probability of refusing to make a decision. The structural-functional organization of interaction between sensory and cognitive processes in learning and keeping the information in the operative memory, is discussed in association with the control of motivation and attention system and the role of the cortex associative areas.


Subject(s)
Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Learning/physiology , Memory/physiology , Alzheimer Disease/chemically induced , Alzheimer Disease/psychology , Animals , Disease Models, Animal , Macaca mulatta , Neurotoxins
6.
Neurosci Lett ; 310(1): 21-4, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11524148

ABSTRACT

Overproduction of the peptide amyloid beta (Abeta) is thought to be a critical pathogenetic event in Alzheimer's disease (AD). Decreasing A production may therefore slow or halt the progression of AD. In vitro work has indicated that cholinergic muscarinic receptor agonists may reduce cellular production of Abeta. Here we show that systemic administration of physostigmine, an acetylcholinesterase inhibitor, lowers Abeta levels in vivo. Guinea pigs treated for 10 days with s.c. physostigmine had levels of cortical AbetaN-40 and N-42 which were 57% and 72%, respectively, of those in control animals. Levels of cortical beta-amyloid precursor protein were not significantly affected by drug treatment. These results suggest that cholinergic therapy may affect the course of AD by limiting Abeta accumulation.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Chemistry/drug effects , Cholinesterase Inhibitors/pharmacology , Physostigmine/pharmacology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinesterase Inhibitors/administration & dosage , Female , Guinea Pigs , Injections, Subcutaneous , Physostigmine/administration & dosage
7.
J Biol Chem ; 276(16): 12991-8, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11152675

ABSTRACT

We have undertaken an integrated chemical and morphological comparison of the amyloid-beta (Abeta) molecules and the amyloid plaques present in the brains of APP23 transgenic (tg) mice and human Alzheimer's disease (AD) patients. Despite an apparent overall structural resemblance to AD pathology, our detailed chemical analyses revealed that although the amyloid plaques characteristic of AD contain cores that are highly resistant to chemical and physical disruption, the tg mice produced amyloid cores that were completely soluble in buffers containing SDS. Abeta chemical alterations account for the extreme stability of AD plaque core amyloid. The corresponding lack of post-translational modifications such as N-terminal degradation, isomerization, racemization, pyroglutamyl formation, oxidation, and covalently linked dimers in tg mouse Abeta provides an explanation for the differences in solubility between human AD and the APP23 tg mouse plaques. We hypothesize either that insufficient time is available for Abeta structural modifications or that the complex species-specific environment of the human disease is not precisely replicated in the tg mice. The appraisal of therapeutic agents or protocols in these animal models must be judged in the context of the lack of complete equivalence between the transgenic mouse plaques and the human AD lesions.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Brain Chemistry , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/isolation & purification , Animals , Brain/pathology , Chromatography, High Pressure Liquid , Cyanogen Bromide , Humans , Mice , Mice, Transgenic , Molecular Weight , Peptide Fragments/chemistry , Plaque, Amyloid/chemistry , Plaque, Amyloid/pathology , Trypsin
8.
Mol Med ; 7(9): 609-18, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11778650

ABSTRACT

BACKGROUND: High levels of A beta in the cerebral cortex distinguish demented Alzheimer's disease (AD) from nondemented elderly individuals, suggesting that decreased amyloid-beta (A beta) peptide clearance from the brain is a key precipitating factor in AD. MATERIALS AND METHODS: The levels of A beta in brain and plasma as well as apolipoprotein E (ApoE) in brain were investigated by enzyme-linked immunosorbent assay (ELISA) and Western blotting at various times during the life span of the APP23 transgenic (Tg) and control mice. Histochemistry and immunocytochemistry were used to assess the morphologic characteristics of the brain parenchymal and cerebrovascular amyloid deposits and the intracellular amyloid precursor protein (APP) deposits in the APP23 Tg mice. RESULTS: No significant differences were found in the plasma levels of A beta between the APP23 Tg and control mice from 2-20 months of age. In contrast, soluble A beta levels in the brain were continually elevated, increasing 4-fold at 2 months and 33-fold in the APP23 Tg mice at 20 months of age when compared to the control mice. Soluble A beta42 was about 60% higher than A beta40. In the APP23 Tg mice, insoluble A beta40 remained at basal levels in the brain until 9 months and then rose to 680 microg/g cortex by 20 months. Insoluble A beta40 was negligible in non-Tg mice at all ages. Insoluble A beta42 in APP23 Tg mice rose to 60 microg/g cortex at 20 months, representing 24 times the control A beta42 levels. Elevated levels of ApoE in the brain were observed in the APP23 Tg mice at 2 months of age, becoming substantially higher by 20 months. ApoE colocalized with A beta in the plaques. Beta-amyloid precursor protein (betaAPP) deposits were detected within the neuronal cytoplasm from 4 months of age onward. Amyloid angiopathy in the APP23 Tg mice increased markedly with age, being by far more severe than in the Tg2576 mice. CONCLUSIONS: We suggest that the APP23 Tg mouse may develop an earlier blockage in A beta clearance than the Tg2576 mice, resulting in a more severe accumulation of A beta in the perivascular drainage pathways and in the brain. Both Tg mice reflect decreased A beta elimination and as models for the amyloid cascade they are useful to study AD pathophysiology and therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Peptide Fragments/metabolism , Aging/physiology , Alzheimer Disease/etiology , Amyloid beta-Peptides/blood , Animals , Apolipoproteins E/metabolism , Brain/blood supply , Brain/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Peptide Fragments/blood , Time Factors
9.
Biochemistry ; 39(51): 16119-24, 2000 Dec 26.
Article in English | MEDLINE | ID: mdl-11123940

ABSTRACT

Extracellular senile plaques composed predominantly of fibrillar amyloid-beta (Abeta) are a major neuropathological feature of Alzheimer's disease (AD). Genetic evidence and in vivo studies suggest that apolipoprotein E (apoE) may contribute to amyloid clearance and/or deposition. In vitro studies demonstrate that native apoE2 and E3 form an SDS-stable complex with Abeta(1-40), while apoE4 forms little such complex. Our current work extends these observations by presenting evidence that apoE3 also binds to Abeta(1-42) and with less avidity to modified species of the peptide found in senile plaque cores. These modified peptides include a form that originates at residue 3-Glu as pyroglutamyl and another with isomerization at the 1-Asp and 7-Asp positions. In addition, we used binding reactions between apoE3 and various Abeta fragments, as well as binding reactions with apoE3 and Abeta(1-40) plus Abeta fragments as competitors, to identify the domain(s) of Abeta involved in the formation of an SDS-stable complex with apoE3. Residues 13-28 of Abeta appear to be necessary, while complex formation is further enhanced by the presence of residues at the C-terminus of the peptide. These results contribute to our understanding of the biochemical basis for the SDS-stable apoE3/Abeta complex and support the hypothesis that Abeta can be transported in vivo complexed with apoE. This complex may then be cleared from the interstitial space by apoE receptors in the brain or become part of an extracellular amyloid deposit.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Peptide Fragments/metabolism , Amino Acid Substitution , Amyloid beta-Peptides/pharmacology , Apolipoprotein E3 , Apolipoproteins E/pharmacology , Binding, Competitive , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , Macromolecular Substances , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Sodium Dodecyl Sulfate
10.
Mol Med ; 6(5): 430-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10952022

ABSTRACT

BACKGROUND: Amyloid-beta (A beta) accumulates in plaques and as cerebral amyloid angiopathy (CAA) in the brains of both Alzheimer's disease (AD) patients and transgenic A betaPPswe/tg2576 (tg2576) mice. Increasingly, evidence in humans and mice shows this process to be modulated by apolipoprotein E (apoE). MATERIALS AND METHODS: To explore this relationship, we measured apoE and A beta levels in brains of tg2576 mice and controls at intervals between 2 and 20 months. In addition, A beta concentrations in plasma and muscle of these animals were also quantified. RESULTS: Quite strikingly, we found that the amount of tg2576 mice brain apoE was elevated by an average of 45%, relative to the control mice from 2 months on. The level of brain apoE soared after 14 months to almost 60% greater than the level found in control mice. A beta concentrations in brains before 9 months were less than 2 ng/mg of protein, but by 14 months concentrations rose to 8.7 ng/mg, and by 20 months to 47 ng/mg. In plasma, we noted that the levels of A beta in tg2576 mice declined from above 30 ng/ml prior to 12 months to 14 ng/ml by 14 months. Histology showed that A beta plaques and CAA began to be discernible in the tg2576 mice at about 9 and 20 months of age, respectively. CONCLUSIONS: ApoE was immunocytochemically detected in neuritic plaques that were positive for thioflavine-S. We suggest that the elevation of brain apoE in tg2576 mice participates in an age-related dysregulation of A beta clearance and signals the start of A beta sequestration during the time of cognitive dysfunction.


Subject(s)
Aging , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Apolipoproteins E/genetics , Brain/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid/analysis , Amyloid/metabolism , Animals , Apolipoproteins E/metabolism , Brain/growth & development , Brain/pathology , Humans , Learning Disabilities/etiology , Memory Disorders/etiology , Mice , Mice, Transgenic
11.
Biochim Biophys Acta ; 1502(1): 31-43, 2000 Jul 26.
Article in English | MEDLINE | ID: mdl-10899429

ABSTRACT

In this chapter, we attempt to analyze the evolution of the amyloid-beta (Abeta) molecular structure from its inception as part of the Abeta precursor protein to its release by the secretases and its extrusion from membrane into an aqueous environment. Biophysical studies suggest that the Abeta peptide sustains a series of transitions from a molecule rich in alpha-helix to a molecule in which beta-strands prevail. It is proposed that initially the extended C-termini of two opposing Abeta dimers form an antiparallel beta-sheet and that the subsequent addition of dimers generates a helical Abeta protofilament. Two or more protofilaments create a strand in which the hydrophobic core of the beta-sheets is shielded from the aqueous environment by the N-terminal polar domains of the Abeta dimers. Once the nucleation has occurred, the Abeta filament grows in length by the addition of dimers or tetramers.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/chemistry , Peptides/chemistry , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/chemistry , Cell Membrane/chemistry , Dimerization , Endopeptidases/chemistry , Humans , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Models, Molecular , Molecular Sequence Data , Molecular Structure , Neurofibrillary Tangles/chemistry , Oligopeptides/chemistry , X-Ray Diffraction
12.
Cell Mol Biol (Noisy-le-grand) ; 46(4): 777-83, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10875439

ABSTRACT

Abeta derived from amyloid plaques of Alzheimer's disease-affected brain contain several oxidative posttranslational modifications. In this study we have characterized the amino acid content of human amyloid-derived Abeta and compared it with that of human synthetic Abeta subjected to metal-catalyzed oxidation. Human amyloid derived Abeta has an increased content of arginine (46%) and glutamate/glutamine residues (28%), but a decreased content of histidine residues (-32%) as compared to the expected amino acid content. Incubation of synthetic human Abeta with Cu(II), but not Fe(III), in the presence of H2O2 similarly induced a decrease in histidine residues (-79%), but also a decrease in tyrosine residues (-28%). Our results suggest that histidine and tyrosine are most vulnerable to metal mediated oxidative attack, consistent with our earlier findings that Cu coordinated via histidine residues is redox competent. Our results suggest that the loss of histidine residues in human amyloid-derived Abeta may be a result of Cu oxidation, and that unidentified post-translational mechanisms operate to modify other amino acids of Abeta in vivo.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Copper/chemistry , Oxygen/metabolism , Peptide Fragments/chemistry , Amino Acids/chemistry , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/metabolism , Arginine/chemistry , Catalysis , Chromatography, High Pressure Liquid , Copper/metabolism , Glutamic Acid/chemistry , Glutamine/chemistry , Histidine/chemistry , Humans , Hydrogen Peroxide/metabolism , Iron/metabolism , Oxidation-Reduction , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Time Factors , Tyrosine/chemistry
13.
Ann N Y Acad Sci ; 903: 110-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10818495

ABSTRACT

Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of beta-amyloid (A beta) peptides in the walls of arteries both in the cortex and meninges. Here, we test the hypothesis that CAA results from the progressive accumulation of A beta in the perivascular interstitial fluid drainage pathways of the brain. Experimental studies have shown that interstitial fluid (ISF) from the rat brain flows along periarterial spaces to join the cerebrospinal fluid (CSF) to drain to cervical lymph nodes. Such lymphatic drainage plays a key role in B-cell and T-cell mediated immunity of the brain. Anatomical studies have defined periarterial ISF drainage pathways in the human brain that are homologous with the lymphatic pathways in the rat brain but are largely separate from the CSF. Periarterial channels in the brain in man are in continuity with those of leptomeningeal arteries and can be traced from the brain to the extracranial portions of the internal carotid arteries related to deep cervical lymph nodes. The pattern of deposition of A beta in senile plaques and in CAA suggests that A beta accumulates in pericapillary and periarterial ISF drainage pathways. A beta could accumulate in CAA due to either (i) increased production of A beta, (ii) reduced solubility of A beta peptides, or (iii) impedance of drainage of A beta along periarterial ISF drainage pathways within the brain and leptomeninges due to aging factors in cerebral arteries. Elucidation of factors that reduce elimination of A beta via perivascular drainage pathways may lead to their rectification and to new strategies for treatment of Alzheimer's disease.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Brain/physiopathology , Cerebral Amyloid Angiopathy/physiopathology , Extracellular Space/physiology , Lymph/physiology , Animals , Brain/blood supply , Humans , Models, Neurological , Plaque, Amyloid/physiology , Rats
14.
Ann N Y Acad Sci ; 903: 118-22, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10818496

ABSTRACT

The increased risk for Alzheimer's Disease (AD) associated with traumatic brain injury (TBI) suggests that environmental insults may influence the development of this age-related dementia. Recently, we have shown that the levels of the beta-amyloid peptide (A beta 1-42) increase in the cerebrospinal fluid (CSF) of patients after severe brain injury and remain elevated for some time after the initial event. The relationships of elevated A beta with markers of blood-brain barrier (BBB) disruption, inflammation, and nerve cell or axonal injury were evaluated in CSF samples taken daily from TBI patients. This analysis reveals that the rise in A beta 1-42 is best correlated with possible markers of neuronal or axonal injury, the cytoskeletal protein tau, neuron-specific enolase (NSE), and apolipoprotein E (ApoE). Similar or better correlations were observed between A beta 1-40 and the three aforementioned markers. These results imply that the degree of brain injury may play a decisive role in determining the levels of A beta 1-42 and A beta 1-40 in the CSF of TBI patients. Inflammation and alterations in BBB may play lesser, but nonetheless significant, roles in determining the A beta level in CSF after brain injury.


Subject(s)
Acute-Phase Proteins/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Brain Injuries/cerebrospinal fluid , Cytokines/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Alzheimer Disease/epidemiology , Amyloid beta-Protein Precursor/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Blood-Brain Barrier , Brain Injuries/complications , Cohort Studies , Humans , Interleukin-6/cerebrospinal fluid , Interleukin-8/cerebrospinal fluid , Phosphopyruvate Hydratase/cerebrospinal fluid , Risk Factors , Transforming Growth Factor beta/cerebrospinal fluid , Tumor Necrosis Factor-alpha/cerebrospinal fluid , tau Proteins/cerebrospinal fluid
15.
Ann N Y Acad Sci ; 903: 366-73, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10818527

ABSTRACT

Selective destruction of the cholinergic nucleus basalis magnocellularis (nbm) in the rabbit by the p75 neurotrophin receptor (NTR) immunoglobulin G (IgG) complexed to the toxin saporin leads to the deposition of amyloid-beta (A beta) in and around cerebral blood vessels. In some instances, the perivascular A beta resemble the diffuse deposits observed in Alzheimer's disease (AD). We propose that cortical cholinergic deprivation results, among other perturbations, in the loss of vasodilation mediated by acetylcholine. In addition to a dysfunctional cerebral blood flow, alterations in vascular chemistry affecting endothelial and smooth muscle cells may result in cerebral hypoperfusion and a breached blood-brain barrier (BBB). The selective removal of the rabbit nbm and A beta accumulation may serve as an important nontransgenic, and more physiological, model for the testing of pharmacological and immunological agents designed to control the deposition and the deleterious effects of A beta in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Cortex/pathology , Cerebrovascular Circulation/physiology , Immunotoxins/toxicity , Microcirculation/pathology , N-Glycosyl Hydrolases , Plant Proteins/toxicity , Receptors, Nerve Growth Factor/physiology , Acetylcholine/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/blood , Animals , Antibodies, Monoclonal , Basal Nucleus of Meynert/pathology , Basal Nucleus of Meynert/physiopathology , Blood-Brain Barrier , Cerebral Cortex/blood supply , Cerebral Cortex/physiopathology , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Cerebrovascular Circulation/drug effects , Denervation , Functional Laterality , Immunoglobulin G , Injections, Intraventricular , Microcirculation/drug effects , Microcirculation/metabolism , Plant Proteins/administration & dosage , Rabbits , Receptors, Nerve Growth Factor/antagonists & inhibitors , Regional Blood Flow , Ribosome Inactivating Proteins, Type 1 , Saporins
16.
J Neurosci ; 20(10): 3606-11, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10804202

ABSTRACT

Many neurodegenerative diseases are associated with the abnormal sequestration of disease-specific proteins in the brain, but the events that initiate this process remain unclear. To determine whether the deposition of the beta-amyloid peptide (Abeta), a key pathological feature of Alzheimer's disease (AD), can be induced in vivo, we infused dilute supernatants of autopsy-derived neocortical homogenates from Alzheimer's patients unilaterally into the hippocampus and neocortex of 3-month-old beta-amyloid precursor protein (betaAPP)-transgenic mice. Up to 4 weeks after the infusion there was no Abeta-deposition in the brain; however, after 5 months, the AD-tissue-injected hemisphere of the transgenic mice had developed profuse Abeta-immunoreactive senile plaques and vascular deposits, some of which were birefringent with Congo Red. There was limited deposition of diffuse Abeta also in the brains of betaAPP-transgenic mice infused with tissue from an age-matched, non-AD brain with mild beta-amyloidosis, but none in mice receiving extract from a young control case. Abeta deposits also were not found in either vehicle-injected or uninjected transgenic mice or in any nontransgenic mice. The results show that cerebral beta-amyloid can be seeded in vivo by a single inoculation of dilute AD brain extract, demonstrating a key pathogenic commonality between beta-amyloidosis and other neurodegenerative diseases involving abnormal protein polymerization. The paradigm can be used to clarify the conditions that initiate in vivo beta-amyloidogenesis in the brain and may yield a more authentic animal model of Alzheimer's disease and other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Adult , Aged , Aged, 80 and over , Amyloid beta-Peptides/immunology , Animals , Antigen-Antibody Reactions , Cerebral Arteries/pathology , Coloring Agents , Congo Red , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/pathology , Female , Hippocampus/pathology , Humans , Injections, Intraventricular , Male , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/pathology
17.
J Neuropathol Exp Neurol ; 59(4): 308-13, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10759186

ABSTRACT

Effective therapeutic intervention in Alzheimer disease (AD) will be most effective if it is directed at early events in the pathogenic sequence. The cholinergic deficit may be such an early event. In the present study, the brains of 26 subjects who had no history of cognitive loss and who were in early histopathologic stages of AD (average Braak stage less than II) were examined at autopsy to determine whether a cortical cholinergic decrement was associated with Abeta concentration or deposition. In the superior frontal and inferior temporal gyri, the choline acetyltransferase (ChAT) activity of plaque-containing cases was significantly decreased (p < 0.05, unpaired, two-tailed t-tests), measuring 70.9% and 79.5%, respectively, relative to plaque-free cases. In the inferior temporal gyrus, Spearman's rank correlation analysis showed that ChAT activity had a significant inverse correlation with Abeta concentration (p = 0.075; r = -0.3552). The results indicate that the cholinergic deficit is established at an early histopathologic stage of AD, before the onset of clinical symptoms.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Choline O-Acetyltransferase/deficiency , Aged , Aged, 80 and over , Choline O-Acetyltransferase/metabolism , Disease Progression , Frontal Lobe/enzymology , Frontal Lobe/pathology , Gyrus Cinguli/enzymology , Gyrus Cinguli/pathology , Humans , Middle Aged , Plaque, Amyloid/pathology , Severity of Illness Index , Temporal Lobe/enzymology , Temporal Lobe/pathology
18.
Neurosci Lett ; 283(1): 9-12, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10729621

ABSTRACT

Brain deposition of the amyloid beta-peptide (Abeta) is a critical step in the pathogenesis of Alzheimer's disease (AD) and human cerebral amyloid angiopathy (CAA). A small fraction of AD and CAA cases are caused by gene mutations leading to increased production and deposition of Abeta, but for the majority, there is no known direct genetic cause. We have hypothesized that Abeta deposition in these sporadic cases occurs as a result of cortical cholinergic deafferentation. Here we show that cortical cholinergic deafferentation, induced in rabbits by a selective immunotoxin, leads to Abeta deposition in cerebral blood vessels and perivascular neuropil. Biochemical measurements confirmed that lesioned animals had 2.5- and 8-fold elevations of cortical Abeta40 and Abeta42, respectively. Cholinergic deafferentation may be one factor that can contribute to Abeta deposition.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Nerve Degeneration/chemically induced , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/drug effects , Animals , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/pathology , Basal Nucleus of Meynert/physiopathology , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Cortex/cytology , Choline O-Acetyltransferase/drug effects , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/pathology , Denervation , Disease Models, Animal , Immunotoxins/pharmacology , Neurons/drug effects , Neurons/pathology , Neurotoxins/pharmacology , Rabbits
19.
Am J Pathol ; 156(3): 797-805, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10702395

ABSTRACT

The levels of amyloid-beta40 (Abeta40) and Abeta42 peptides were quantified in temporalis muscles and brain of neuropathologically diagnosed Alzheimer disease (AD) and of nondemented individuals. This was achieved by using a novel analytical approach consisting of a combination of fast-performance liquid chromatographic (FPLC) size exclusion chromatography developed under denaturing conditions and europium immunoassay on the 4.0- to 4.5-kd fractions. In the temporalis muscles of the AD and nondemented control groups, the average values for Abeta42 were 15.7 ng/g and 10.2 ng/g (P = 0.010), and for Abeta40 they were 37.8 ng/g and 29.8 ng/g (P = 0.067), respectively. Multiple regression analyses of the AD and control combined populations indicated that 1) muscle Abeta40 and muscle Abeta42 levels were correlated with each other (P < 0.001), 2) muscle Abeta40 levels were positively correlated with age (P = 0. 036), and 3) muscle Abeta42 levels were positively correlated with Braak stage (P = 0.042). Other forms of the Abeta peptide were discovered by mass spectrometry, revealing the presence of Abeta starting at residues 1, 6, 7, 9, 10, and 11 and ending at residues 40, 42, 44, 45, and 46. It is possible that in AD the skeletal muscle may contribute to the elevated plasma pool of Abeta and thus indirectly to the amyloid deposits of the brain parenchyma and cerebral blood vessels. The increased levels of Abeta in the temporalis muscles of AD patients suggest that alterations in AbetaPP and Abeta metabolism may be manifested in peripheral tissues.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Temporal Muscle/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/isolation & purification , Blotting, Western , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Female , Humans , Immunoenzyme Techniques , Male , Mass Spectrometry , Middle Aged , Peptide Fragments/isolation & purification
20.
Biochem Biophys Res Commun ; 268(3): 750-6, 2000 Feb 24.
Article in English | MEDLINE | ID: mdl-10679277

ABSTRACT

Amyloid beta peptides are bound rapidly in the plasma complicating an accurate assessment of their in vivo abundance by immunoassay procedures. The extent of Abeta immunoassay interference was used to estimate the Abeta binding capacity of purified plasma proteins, erythrocytes and whole plasma. Human serum albumin bound Abeta peptides rapidly with a 1:1 stoichiometry and at physiological concentrations was capable of binding over 95% of an input of 5 ng/ml Abeta. Purified alpha2-macroglobulin was able to bind Abeta peptides and at physiological concentration bound 73% of 5 ng/ml of Abeta. Erythrocytes also sequestered the Abeta peptides, showing a preference for binding Abeta 1-42. Incubation of 5 ng/ml of Abeta in plasma revealed that about 30% of the peptides were still detectable by immunoassay, presumably reflecting the binding of Abeta peptides with albumin and other plasma molecules. Thus, our studies reveal that both the soluble and formed elements of the blood are capable of sequestering Abeta peptides. To avoid underestimating plasma Abeta values, we employed an improved column chromatography method under denaturing conditions to liberate Abeta from its associations with plasma proteins. Quantification of Abeta 40 and 42 levels in plasma from both normal and AD individuals after chromatography showed a large overlap between AD and control groups, despite the very large pool of Abeta present in the AD brains. The potential origins of the plasma Abeta pool are discussed.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Blood Proteins/metabolism , Erythrocytes/metabolism , Aged , Aged, 80 and over , Blood Chemical Analysis , Case-Control Studies , Female , Humans , Immunoassay , In Vitro Techniques , Male , Middle Aged , Peptide Fragments/blood , Protein Binding , Serum Albumin/metabolism , alpha-Macroglobulins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...