Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Brain Res ; 1620: 153-68, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-25966615

ABSTRACT

Vascular and glial involvement in the development of neurodegenerative disorders, such as Alzheimer's disease (AD), and age-related brain vulnerabilities have been suggested. Therefore, we sought to: (i) investigate which vascular and glial events are evident in ageing and/or AD, (ii) to establish the temporal evolution of vascular and glial changes in AD-like and wild-type (WT) mice and (iii) to relate them to amyloid-ß (Aß) peptide accumulation. We examined immunohistochemically hippocampi and cortex from APP/PS1dE9 and WT C57BL/6 mice along ageing and disease progression (young-adulthood, middle- and old-age). Ageing resulted in the increase in receptor for advanced glycation endproducts expression, as well as the entrance of thrombin and albumin in hippocampal parenchyma. In contrast, the loss of platelet-derived growth factor receptor-ß (PDGFR-ß) positive cells, in both regions, was only related to AD pathogenesis. Hypovascularization was affected by both ageing and AD in the hippocampus, but resulted from the interaction between both factors in the cortex. Astrogliosis was a result of AD in hippocampus and of both factors in cortex, while microgliosis was associated with fibrillar amyloid plaques in AD-like mice and with the interaction between both factors in each of the studied regions. In sum, these data show that senile plaques precede vascular and glial alterations only in hippocampus, whereas in cortex, vascular and glial alterations, namely the loss of PDGFR-ß-positive cells and astrogliosis, accompanied the first senile plaques. Hence, this study points to vascular and glial events that co-exist in AD pathogenesis and age-related brain vulnerabilities.


Subject(s)
Aging/physiology , Alzheimer Disease/physiopathology , Cerebral Cortex/physiopathology , Hippocampus/physiopathology , Neuroglia/physiology , Aging/pathology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Capillaries/pathology , Capillaries/physiopathology , Cerebral Cortex/pathology , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/pathology , Pericytes/pathology , Pericytes/physiology , Presenilin-1/genetics , Presenilin-1/metabolism
2.
CNS Neurol Disord Drug Targets ; 9(2): 197-206, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20205640

ABSTRACT

Alzheimer's disease (AD) is a progressive, degenerative disorder of the brain and the most common form of dementia among the elderly. As the population grows and lifespan is extended, the number of AD patients will continue to rise. Current clinical therapies for AD provide partial symptomatic benefits for some patients; however, none of them modify disease progression. Amyloid-beta (Abeta) peptide, the major component of senile plaques in AD patients, is considered to play a crucial role in the pathogenesis of AD thereby leading to Abeta as a target for treatment. Abeta immunotherapy has been shown to induce a marked reduction in amyloid burden and an improvement in cognitive function in animal models. Although preclinical studies were successful, the initial human clinical trial of an active Abeta vaccine was halted due to the development of meningoencephalitis in approximately 6% of the vaccinated AD patients. Some encouraging outcomes, including signs of cognitive stabilization and apparent plaque clearance, were obtained in subset of patients who generated antibody titers. These promising preliminary data support further efforts to refine Abeta immunotherapy to produce highly effective and safer active and passive vaccines for AD. Furthermore, some new human clinical trials for both active and passive Abeta immunotherapy are underway. In this review, we will provide an update of Abeta immunotherapy in animal models and in human beings, as well as discuss the possible mechanisms underlying Abeta immunotherapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Vaccines/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Brain/drug effects , Immunotherapy/methods , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Alzheimer Vaccines/adverse effects , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Brain/immunology , Brain/physiopathology , Clinical Trials as Topic/statistics & numerical data , Disease Models, Animal , Humans , Immunotherapy/trends , Mice , Mice, Transgenic/genetics , Mice, Transgenic/immunology , Mice, Transgenic/metabolism , Plaque, Amyloid/drug effects , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Vaccination/methods , Vaccination/trends
3.
Proc Natl Acad Sci U S A ; 106(29): 12145-50, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19581601

ABSTRACT

A number of distinct beta-amyloid (Abeta) variants or multimers have been implicated in Alzheimer's disease (AD), and antibodies recognizing such peptides are in clinical trials. Humans have natural Abeta-specific antibodies, but their diversity, abundance, and function in the general population remain largely unknown. Here, we demonstrate with peptide microarrays the presence of natural antibodies against known toxic Abeta and amyloidogenic non-Abeta species in plasma samples and cerebrospinal fluid of AD patients and healthy controls aged 21-89 years. Antibody reactivity was most prominent against oligomeric assemblies of Abeta and pyroglutamate or oxidized residues, and IgGs specific for oligomeric preparations of Abeta1-42 in particular declined with age and advancing AD. Most individuals showed unexpected antibody reactivities against peptides unique to autosomal dominant forms of dementia (mutant Abeta, ABri, ADan) and IgGs isolated from plasma of AD patients or healthy controls protected primary neurons from Abeta toxicity. Aged vervets showed similar patterns of plasma IgG antibodies against amyloid peptides, and after immunization with Abeta the monkeys developed high titers not only against Abeta peptides but also against ABri and ADan peptides. Our findings support the concept of conformation-specific, cross-reactive antibodies that may protect against amyloidogenic toxic peptides. If a therapeutic benefit of Abeta antibodies can be confirmed in AD patients, stimulating the production of such neuroprotective antibodies or passively administering them to the elderly population may provide a preventive measure toward AD.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Antibodies/immunology , Neuroprotective Agents/immunology , Peptides/immunology , Aging/drug effects , Alzheimer Disease/blood , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Antibodies/blood , Antibodies/cerebrospinal fluid , Cytoprotection/drug effects , Dementia/complications , Dementia/immunology , Disease Progression , Genes, Dominant , Immunization , Immunoglobulin G/blood , Mice , Molecular Weight , Neurons/cytology , Neurons/drug effects , Peptides/chemistry , Primates/immunology , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary
4.
Neuroscience ; 150(2): 386-95, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17945434

ABSTRACT

Alpha-secretase (alpha-secretase), cleaves the amyloid precursor protein (APP) within the amyloid-beta (Abeta) sequence, resulting in the release of a secreted fragment of APP (alphaAPPs) and precluding Abeta generation. We investigated the effects of the acetylcholinesterase inhibitor, huperzine A (Hup A), on APP processing and Abeta generation in human neuroblastoma SK-N-SH cells overexpressing wild-type human APP695. Hup A dose-dependently (0-10 microM) increased alphaAPPs release. Therefore, we evaluated two alpha-secretase candidates, a disintegrin and metalloprotease (ADAM) 10 and ADAM17 in Hup A-induced non-amyloidogenic APP metabolism. Hup A enhanced the level of ADAM10, and the inhibitor of tumor necrosis factor-alpha converting enzyme (TACE)/ADAM17 inhibited the Hup A-induced rise in alphaAPPs levels, further suggesting Hup A directed APP metabolism toward the non-amyloidogenic alpha-secretase pathway. Hup A had no effect on Abeta generation in this cell line. The steady-state levels of full-length APP and cell viability were unaffected by Hup A. Alpha-APPs release induced by Hup A treatment was significantly reduced by muscarinic acetylcholine receptor antagonists (particularly by an M1 antagonist), protein kinase C (PKC) inhibitors, GF109203X and calphostin C, and the mitogen-activated kinase kinase (MEK) inhibitors, U0126 and PD98059. Furthermore, Hup A markedly increased the phosphorylation of p44/p42 mitogen-activated protein (MAP) kinase, which was blocked by treatment with U0126 and PD98059. In addition, Hup A inhibited acetylcholinesterase activity by 20% in neuroblastoma cells. Our results indicate that the activation of muscarinic acetylcholine receptors, PKC and MAP kinase may be involved in Hup A-induced alphaAPPs secretion in neuroblastoma cells and suggest multiple pharmacological mechanisms of Hup A regarding the treatment of Alzheimer's disease (AD).


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/drug effects , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Sesquiterpenes/pharmacology , ADAM Proteins/drug effects , ADAM Proteins/metabolism , ADAM10 Protein , ADAM17 Protein , Alkaloids , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cholinesterase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System/physiology , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Muscarinic Antagonists/pharmacology , Neuroblastoma , Neurons/metabolism , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism
5.
Am J Pathol ; 158(4): 1345-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11290552

ABSTRACT

Mutations in the amyloid precursor protein (APP) and presenilin-1 and -2 genes (PS-1, -2) cause Alzheimer's disease (AD). Mice carrying both mutant genes (PS/APP) develop AD-like deposits composed of beta-amyloid (Abeta) at an early age. In this study, we have examined how Abeta deposition is associated with immune responses. Both fibrillar and nonfibrillar Abeta (diffuse) deposits were visible in the frontal cortex by 3 months, and the amyloid load increased dramatically with age. The number of fibrillar Abeta deposits increased up to the oldest age studied (2.5 years old), whereas there were less marked changes in the number of diffuse deposits in mice over 1 year old. Activated microglia and astrocytes increased synchronously with amyloid burden and were, in general, closely associated with deposits. Cyclooxygenase-2, an inflammatory response molecule involved in the prostaglandin pathway, was up-regulated in astrocytes associated with some fibrillar deposits. Complement component 1q, an immune response component, strongly colocalized with fibrillar Abeta, but was also up-regulated in some plaque-associated microglia. These results show: i) an increasing proportion of amyloid is composed of fibrillar Abeta in the aging PS/APP mouse brain; ii) microglia and astrocytes are activated by both fibrillar and diffuse Abeta; and iii) cyclooxygenase-2 and complement component 1q levels increase in response to the formation of fibrillar Abeta in PS/APP mice.


Subject(s)
Alzheimer Disease/metabolism , Amyloidosis/metabolism , Inflammation Mediators/metabolism , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloidosis/genetics , Amyloidosis/pathology , Amyloidosis/physiopathology , Animals , Complement C1q/metabolism , Cyclooxygenase 2 , Isoenzymes/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic/genetics , Neuroglia/physiology , Presenilin-1 , Presenilin-2 , Prostaglandin-Endoperoxide Synthases/metabolism , Tissue Distribution
6.
DNA Cell Biol ; 20(11): 705-11, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11788048

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disease for which there is currently no effective prevention or treatment. The prediction that the number of U.S. patients with AD will triple to approximately 14 million over the next 50 years underscores the urgent need to explore novel therapeutic strategies for AD. The beta-amyloid protein (Abeta) accumulation and accompanying inflammation appear to play key roles in initiating the neuronal degeneration that underlies the signs and symptoms of AD. Interventions geared toward reducing Abeta accumulation and inflammatory responses should delay or prevent the onset of the clinical disease. Recently, several research groups, including ours, have shown that vaccination with Abeta results in a significant lowering of the Abeta burden in the brains of APP transgenic mice and, in some studies, improvement in their cognitive deficits. Our study described a novel approach, namely mucosal (intranasal) Abeta vaccination. Precisely how Abeta vaccination chronically lowers Abeta levels and reduces Abeta-associated pathology remains unclear. Here, we provide an overview of these studies, with particular emphasis on our work with intranasal Abeta vaccination. Examples of other intranasal vaccines and mucosal adjuvants are presented. Taken together, these data have implications for the future development of an intranasal Abeta vaccine for humans.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/administration & dosage , Adjuvants, Immunologic/therapeutic use , Administration, Intranasal , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Antibody Formation , Brain/metabolism , Disease Models, Animal , Mice , Mice, Transgenic
7.
Ann Neurol ; 48(4): 567-79, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11026440

ABSTRACT

Progressive cerebral deposition of amyloid-beta (Abeta) peptide, an early and essential feature of Alzheimer's disease (AD), is accompanied by an inflammatory reaction marked by microgliosis, astrocytosis, and the release of proinflammatory cytokines. Mucosal administration of disease-implicated proteins can induce antigen-specific anti-inflammatory immune responses in mucosal lymphoid tissue which then act systemically. We hypothesized that chronic mucosal administration of Abeta peptide might induce an anti-inflammatory process in AD brain tissue that could beneficially affect the neuropathological findings. To test this hypothesis, we treated PDAPP mice, a transgenic line displaying numerous neuropathological features of AD, between the ages of approximately 5 and approximately 12 months with human Abeta synthetic peptide mucosally each week. We found significant decreases in the cerebral Abeta plaque burden and Abeta42 levels in mice treated intranasally with Abeta peptide versus controls treated with myelin basic protein or left untreated. This lower Abeta burden was associated with decreased local microglial and astrocytic activation, decreased neuritic dystrophy, serum anti-Abeta antibodies of the IgG1 and IgG2b classes, and mononuclear cells in the brain expressing the anti-inflammatory cytokines interleukin-4, interleukin-10, and tumor growth factor-beta. Our results demonstrate that chronic nasal administration of Abeta peptide can induce an immune response to Abeta that decreases cerebral Abeta deposition, suggesting a novel mucosal immunological approach for the treatment and prevention of AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid/analysis , Brain/drug effects , Brain/pathology , Disease Models, Animal , Administration, Intranasal , Analysis of Variance , Animals , Brain Chemistry , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice
8.
Neurosci Lett ; 290(2): 93-6, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10936685

ABSTRACT

To clarify (1) the localization of flotillins in human brain tissue and (2) the relationship between senile plaque formation and flotillins localization, we performed Western blotting and immunohistochemical analysis. Flotillins 1 and 2 were shown as 48 and 42 kDa bands, respectively, in human brain. The cerebral cortex showed a broad band-like labeling in entire layers. Flotillins were abundant in pyramidal neurons and astrocytes in the white matter. The intensity of the band-like labeling throughout the cortex became stronger with the development of senile plaque formation, and strongest in Alzheimer's disease. These findings suggest that flotillins are associated with progression of Alzheimer pathology.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Brain/pathology , Brain/physiopathology , Membrane Proteins/metabolism , Adult , Aged , Astrocytes/metabolism , Astrocytes/pathology , Cerebellum/pathology , Cerebellum/physiopathology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Disease Progression , Humans , Middle Aged , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
9.
Am J Pathol ; 156(2): 489-99, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666378

ABSTRACT

The complement system constitutes a series of enzymatic steps involved in the inflammatory response and is activated in Alzheimer's disease (AD). Using Down's syndrome (DS) brains as a temporal model for the progression of AD, we examined components of the complement cascade and their relationship to other principal events in AD pathology: Abeta42 deposition, neuritic changes, neurofibrillary tangles (NFTs), and gliosis (reactive astrocytes, activated microglia). Adjacent sections of frontal cortex from 24 DS subjects ranging in age from 12 to 73 years were immunohistochemically examined for immunoreactivity (IR) of classical complement proteins (Clq and C3), markers indicating activation of complement (C4d and C5b-9), the complement inhibitor apolipoprotein J (apo J), and markers of AD neuropathology. Abeta42-labeled diffuse plaques were first detected in a 12-year-old DS subject and were not labeled by any of the complement antibodies. Colocalization of Abeta42 with Clq, C3, C4d, and/or apo J was first detected in compacted plaques in the brain of a 15-year-old DS patient with features of mature AD pathology, such as reactive astrocytes, activated microglia, dystrophic neurites, and a few NFTs. IR for C4d and C5b-9 (membrane attack complex, MAC) was observed in small numbers of plaque-associated dystrophic neurites and in focal regions of pyramidal neurons in this 15-year-old. The only other young (

Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/metabolism , Complement C4b , Complement System Proteins/metabolism , Down Syndrome/complications , Down Syndrome/metabolism , Molecular Chaperones , Plaque, Amyloid/metabolism , Adolescent , Adult , Aged , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Child , Clusterin , Complement C4/metabolism , Complement Membrane Attack Complex/metabolism , Disease Progression , Down Syndrome/pathology , Glycoproteins/metabolism , Humans , Middle Aged , Peptide Fragments/metabolism , Time Factors
11.
Am J Pathol ; 155(1): 29-37, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10393833

ABSTRACT

Novel plaque-like "AMY" lesions were recently described in the brains of patients with Alzheimer's disease (AD). Using three Abeta antibodies, we now document the co-occurrence of AMY immunoreactivity (IR) with amyloid beta-peptide (Abeta) in the large majority of plaques in AD brain. AMY IR was detected in many compacted plaques, whereas its co-localization with early, diffuse Abeta deposits was rare. AMY IR overlapped considerably or fully with Abeta and, in more severely affected AD brains, decorated the periphery of some plaques. In a temporal series of 29 Down syndrome (DS) brains from patients aged 12 to 73 years, the earliest AMY IR was detected in some plaques at age 15, following the earliest appearance of Abeta plaques (age 12 years), and then accrued within a subset of Abeta deposits, namely, the more spherical, compacted plaques. Brains from DS patients 29 years and older showed AMY staining in many Abeta plaques, as seen in AD. Brains from eight monkeys aged 17 to 34 years and thirty APP transgenic mice aged 8 to 20 months showed Abeta IR but no AMY IR. We conclude that AMY IR represents an amyloid-associated antigen that co-deposits in most but not all Abeta plaques in AD and DS and that accumulation of the AMY antigen follows Abeta deposition in plaques.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/immunology , Antigens/metabolism , Down Syndrome/metabolism , Plaque, Amyloid/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aging/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Child , Haplorhini , Humans , Mice , Mice, Transgenic/genetics , Middle Aged , Reference Values
12.
J Med Chem ; 42(15): 2805-15, 1999 Jul 29.
Article in English | MEDLINE | ID: mdl-10425090

ABSTRACT

The definitive diagnosis of Alzheimer's disease (AD) requires the detection of amyloid plaques in postmortem brain. Although the amount of fibrillar amyloid roughly correlates with the severity of symptoms at the time of death, the temporal relationship between amyloid deposition, neuronal loss, and cognitive decline is unclear. To elucidate this relationship, a noninvasive, practical method for the quantitation of brain amyloid deposition is required. We describe herein the initial stages of a strategy to accomplish this goal by single photon computed tomographic imaging. The amyloid-binding dye Congo Red was modified to allow its conjugation to the monoamine-monoamide bis(thiol) ligand. This ligand complexes technetium(V) in its neutral oxo form. A biphenyl-containing building block was conjugated to the protected ligand, and the product was coupled to the relevant aromatic compounds. Rhenium oxo complexes, which are isosteric, but nonradioactive, analogues of the potential imaging agent technetium oxo complexes, were synthesized. These complexes bound to Abeta amyloid fibrils produced in vitro and stained amyloid plaques and vascular amyloid in AD brain sections.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Organometallic Compounds/chemical synthesis , Peptide Fragments/metabolism , Rhenium , Coloring Agents/metabolism , Congo Red/metabolism , Humans , In Vitro Techniques , Organometallic Compounds/metabolism , Organotechnetium Compounds/chemical synthesis , Organotechnetium Compounds/metabolism , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/metabolism , Staining and Labeling , Tomography, Emission-Computed, Single-Photon
13.
Invest Ophthalmol Vis Sci ; 40(1): 168-74, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888440

ABSTRACT

PURPOSE: To study in an infant rat model of retinopathy of prematurity, the rod photoreceptors, which are known to have attenuated photoresponses. METHODS: Rhodopsin was extracted from whole retinas, the thickness of the rod outer segment (ROS) layer was measured, large phagosomes were counted, and the ROS ultrastructure was examined in the retinas of oxygen-exposed and control rats, ages 13 and 18 days. Rhodopsin absorbances in the ROS were measured by microspectrophotometry at age 20 days. RESULTS: The rhodopsin content did not differ significantly between the oxygen-exposed and control rats at either 13 or 18 days. The thickness of the ROS layer was equal in 13-day-old oxygen-exposed and control rats; however, at 18 days, the ROS layer was significantly thinner in the oxygen-exposed rats than in the control rats. The number of phagosomes did not vary significantly among the oxygen-exposed and control groups. Opsin immunoreactivity was seen only in the ROS layer in oxygen-exposed and control rats. The ROS were disorganized in oxygen-exposed rats. The rhodopsin absorbances of the oxygen-exposed ROS were significantly more variable and higher than in the control rats. CONCLUSIONS: Attenuation of the rod photoresponse parameters does not result simply from shortening of the outer segments and consequent low rhodopsin content. Rather, the structure of the outer segments is altered. A fault in the synthesis of the outer segments, rather than disposal of outer segment discs, is suspected.


Subject(s)
Oxygen/toxicity , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/ultrastructure , Retinopathy of Prematurity/pathology , Animals , Animals, Newborn , Cell Count , Humans , Immunoenzyme Techniques , Infant, Newborn , Microspectrophotometry , Phagosomes/pathology , Rats , Rats, Sprague-Dawley , Retinal Rod Photoreceptor Cells/metabolism , Retinopathy of Prematurity/metabolism , Rhodopsin/metabolism , Rod Cell Outer Segment/ultrastructure , Rod Opsins/metabolism , Vision, Ocular
14.
JAMA ; 277(10): 793-9, 1997 Mar 12.
Article in English | MEDLINE | ID: mdl-9052708

ABSTRACT

OBJECTIVES: To characterize clinical features of a very large pedigree with early-onset Alzheimer disease (AD) in which all affected individuals carry the identical glutamic acid-to-alanine mutation at codon 280 in the presenilin-1 gene. DESIGN: Clinical histories were obtained by patient and family interviews and through medical or civil records. Using standard diagnostic criteria, a case series of 128 individuals was identified, of which 6 have definitive (autopsy-proven) early-onset AD, 93 have probable early-onset AD, and 29 have possible early-onset AD. SETTING: Community based in Antioquia, Colombia. PATIENTS: A population-based sample in which all members of 5 extended families (nearly 3000 individuals) were surveyed. Criteria for inclusion required obtaining sufficient information to categorize the individual as affected. MAIN OUTCOME MEASURES: Age at onset, neuropsychological profile, neurologic history, and examination. RESULTS: The patients had a mean age at onset of 46.8 years (range, 34-62 years). The average interval until death was 8 years. Headache was noted in affected individuals significantly more frequently than in those not affected. The most frequent presentation was memory loss followed by behavior and personality changes and progressive loss of language ability. In the final stages, gait disturbances, seizures, and myoclonus were frequent. CONCLUSIONS: Other than the early onset, this clinical phenotype is indistinguishable from sporadic AD except that affected individuals frequently complained of headache preceding and during the disease. Despite the uniform genetic basis for the disease, there was significant variability in the age at onset, suggesting an important role for environmental factors or genetic modifiers in determining the age at onset.


Subject(s)
Alzheimer Disease/genetics , Membrane Proteins/genetics , Point Mutation , Adult , Age of Onset , Alanine , Alzheimer Disease/diagnosis , Alzheimer Disease/physiopathology , Autopsy , Brain/pathology , Codon , Female , Glutamic Acid , Headache/etiology , Humans , Male , Middle Aged , Neuropsychological Tests , Pedigree , Phenotype , Presenilin-1
15.
J Biol Chem ; 271(52): 33623-31, 1996 Dec 27.
Article in English | MEDLINE | ID: mdl-8969231

ABSTRACT

Down's syndrome (DS) patients show accelerated Alzheimer's disease (AD) neuropathology, which consists of preamyloid lesions followed by the development of neuritic plaques and neurofibrillary tangles. The major constituents of preamyloid and neuritic plaques are amyloid beta (Abeta) peptides. Preamyloid lesions are defined as being Abeta immunoreactive lesions, which unlike neuritic plaque amyloid are Congo red-negative and largely nonfibrillar ultrastructurally. DS patients can develop extensive preamyloid deposits in the cerebellum, without neuritic plaques; hence, DS cerebellums are a source of relatively pure preamyloid. We biochemically characterized the composition of DS preamyloid and compared it to amyloid in the neuritic plaques and leptomeninges in the same patients. We found that Abeta17-42 or p3 is a major Abeta peptide of DS cerebellar preamyloid. This 26-residue peptide is also present in low quantities in neuritic plaques. We suggest that preamyloid can now be defined biochemically as lesions in which a major Abeta peptide is p3.


Subject(s)
Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Down Syndrome/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Benzothiazoles , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Down Syndrome/metabolism , Down Syndrome/pathology , Electrophoresis, Polyacrylamide Gel , Fluorometry , Humans , Mass Spectrometry , Thiazoles/metabolism
16.
Nat Med ; 2(10): 1146-50, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8837617

ABSTRACT

Missense mutations in the presenilin 1 (PS1) gene cause the most common form of dominant early-onset familial Alzheimer's disease (FAD) and are associated with increased levels of amyloid beta-peptides (A beta) ending at residue 42 (A beta 42) in plasma and skin fibroblast media of gene carriers. A beta 42 aggregates readily and appears to provide a nidus for the subsequent aggregation of A beta 40 (ref. 4), resulting in the formation of innumerable neuritic plaques. To obtain in vivo information about how PS1 mutations cause AD pathology at such early ages, we characterized the neuropathological phenotype of four PS1-FAD patients from a large Colombian kindred bearing the codon 280 Glu to Ala substitution (Glu280Ala) PS1 mutation. Using antibodies specific to the alternative carboxy-termini of A beta, we detected massive deposition of A beta 42, the earliest and predominant form of plaque A beta to occur in AD (ref. 6-8), in many brain regions. Computer-assisted quantification revealed a significant increase in A beta 42, but not A beta 40, burden in the brains from 4 PS1-FAD patients compared with those from 12 sporadic AD patients. Severe cerebellar pathology included numerous A beta 42-reactive plaques, many bearing dystrophic neurites and reactive glia. Our results in brain tissue are consistent with recent biochemical evidence of increased A beta 42 levels in PS1-FAD patients and strongly suggest that mutant PS1 proteins alter the proteolytic processing of the beta-amyloid precursor protein at the C-terminus of A beta to favor deposition of A beta 42.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Cerebellum/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Point Mutation , Age of Onset , Aged , Alzheimer Disease/epidemiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain Chemistry , Cerebellum/pathology , Codon/genetics , Female , Humans , Image Processing, Computer-Assisted , Male , Membrane Proteins/physiology , Middle Aged , Nerve Tissue Proteins/physiology , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/pathology , Presenilin-1
17.
Acta Neuropathol ; 92(3): 232-41, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8870824

ABSTRACT

Using immunohistochemistry, we examined the localization of four types of proline-directed kinases in the brains of control rats and in the brains of non-demented aged human subjects, subjects with Alzheimer's disease and those with Down's syndrome. The four kinases were: cyclin-dependent kinase (cdk) 5, a component of tau protein kinase (TPK) II; TPK I/glycogen synthase kinase (GSK)-3 beta; GSK-3 alpha; and mitogen-activated protein kinase (MAPK/ERK2). Each of these kinases has been reported to promote the hyperphosphorylation of tau protein in vitro. The kinases were located essentially in neurons, although the intensity and distribution of labeling varied. Antiserum for cdk5 showed the most preferential and consistent labeling of intraneuronal neurofibrillary tangles (NFT). Antiserum for TPK I/GSK-3 beta also labeled intraneuronal NFT. Double immunolabeling for TPK I/GSK-3 beta and tau 1 showed that TPK I/GSK-3 beta was closely associated with NFT. Antiserum for GSK-3 alpha labeled neurons weakly, and the intensity of labeling did not differ between neurons with and without NFT. Antiserum for MAPK labeled neurons in superficial cortical layers, but NFT appeared in both superficial and deep cortical layers. These findings suggest that cdk5 and TPK I/GSK-3 beta are the critically important kinases for the generation in vivo of hyperphosphorylated tau, the main component of the paired helical filaments in NFT.


Subject(s)
Alzheimer Disease/immunology , Antibody Specificity/immunology , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Cyclin-Dependent Kinases/immunology , Immune Sera/immunology , Neurofibrillary Tangles/immunology , Protein Serine-Threonine Kinases/immunology , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Blotting, Western , Cyclin-Dependent Kinase 5 , Down Syndrome/immunology , Glycogen Synthase Kinase 3 , Humans , Immunohistochemistry , Microtubule-Associated Proteins/immunology , Middle Aged , Mitogen-Activated Protein Kinase 1 , Molecular Sequence Data , Rats , Rats, Wistar
18.
Neurobiol Dis ; 3(1): 16-32, 1996 Feb.
Article in English | MEDLINE | ID: mdl-9173910

ABSTRACT

Patients with trisomy 21 [Down syndrome (DS)] progressively develop amyloid beta-protein (A beta) deposits and then other features of Alzheimer's disease (AD), apparently due to increased gene dosage and thus expression of the beta-amyloid precursor protein. Because the neuropathological phenotype in older DS subjects closely resembles that of AD, the examination of DS brains of increasing age provides a unique model of the progression of AD. Here, we characterized the deposition of several A beta peptides and apolipoprotein E in formalin-fixed brain sections from 29 DS subjects between 3 and 73 years old. Amyloid plaque number and the percentage of cortical area they occupied were quantified by computerized image analysis. A beta ending at amino acid 42 (A beta 42) was the earliest form of A beta deposited in DS cortex. It was observed in 7 of 16 young (3-30 years) subjects, with the earliest deposition occurring at age 12. A beta ending at residue 40 (A beta 40) was not detected until approximately age 30, a time when degenerating neurites around A beta immunoreactive (IR) plaques were first observed, and the frequency of A beta 40 IR plaques then rose with age. Even in old (51-73 years) DS subjects, A beta 42 IR plaques were always more abundant than A beta 40 IR plaques. A beta peptides starting at aspartate 1 or pyroglutamate 3 were detected in small subsets of compacted, neuritic plaques beginning around age 30 and rose with age, the latter species always exceeding the former. Thus, the N-termini of the A beta 42 peptides abundantly deposited in very young DS subjects remain unknown. Apo E was detectable in a small subset of A beta 42 IR plaques beginning at age 12 and rose steadily with age; it clearly followed the deposition of A beta. Our analysis of very young DS brains suggests that amyloid plaque formation begins with A beta 42-ending peptides, and the number and percentage of cortical area of A beta 42 plaques increase very little with advancing age, while other heterogeneous A beta species and Apo E progressively accrue onto plaques containing A beta 42.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism , Down Syndrome/metabolism , Nerve Tissue Proteins/metabolism , Adolescent , Adult , Aged , Aging/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Child , Child, Preschool , Down Syndrome/complications , Down Syndrome/genetics , Down Syndrome/pathology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neurites/pathology , Neurofibrillary Tangles , Peptide Fragments/metabolism , Staining and Labeling , Temporal Lobe/blood supply , Temporal Lobe/metabolism , Temporal Lobe/pathology
19.
Neurobiol Dis ; 3(1): 51-63, 1996 Feb.
Article in English | MEDLINE | ID: mdl-9173912

ABSTRACT

Phospholipase A2 (PLA2) is the key enzyme that initiates the arachidonic acid cascade, which leads to the generation of multiple eicosanoid products. Many of these products are believed to play an important role in the inflammatory process. Activation of PLA2 is observed under pathological conditions where inflammation is present. Cytosolic PLA2 (cPLA2) is activated by very low levels of calcium and is thought to control receptor-mediated eicosanoid production and to participate in intracellular signal transduction processes. In view of the presence of numerous inflammatory mediators and acute phase proteins in the Alzheimer's disease (AD) brain, localization of cPLA2 in AD brain was evaluated and compared to that observed in nonneurologically diseased controls. In this study, a monoclonal antibody raised against cPLA2 was used to immunostain tissue sections of human cerebral cortex. Five AD cases and six neurologically normal cases were evaluated in the occipital cortex and the cerebellum. Two of the AD cases were also examined in other cortical regions. Granular-like staining with anti-cPLA2 was found to be associated with astrocytes in the cortex of both control and AD cases. Colocalization with GFAP confirmed that cPLA2 immunoreactivity is associated almost exclusively with protoplasmic astrocytes. Staining was abolished when sections were labeled with antibody that had been preadsorbed with purified cPLA2. In AD brain, cPLA2 immunoreactive astrocytes were greater in number and more intensely stained than those in control cases. cPLA2 immunoreactivity was virtually absent in the cerebelium of AD and control cases, despite the presence in this region of diffuse amyloid in two AD cases and amyloid angiopathy in a third case. In the cortex, cPLA2 immunoreactive astrocytes were detected in regions that contained numerous A beta deposits. The finding of elevated levels of cPLA2 immunoreactivity in AD brain supports the hypothesis that there is an active inflammatory process occurring in AD.


Subject(s)
Alzheimer Disease/enzymology , Brain/enzymology , Cytosol/enzymology , Isoenzymes/analysis , Nerve Tissue Proteins/analysis , Phospholipases A/analysis , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Arachidonic Acid/metabolism , Astrocytes/enzymology , Biomarkers , Brain/pathology , Cerebellum/enzymology , Cerebellum/pathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Enzyme Activation , Humans , Immunoenzyme Techniques , Inflammation , Phospholipases A2 , Signal Transduction
20.
J Neurobiol ; 29(1): 75-90, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8748373

ABSTRACT

The retinotectal projection is known to be capable of extensive long-term expansion of connections, but it is not known how fast such changes can occur or what triggers sprouting of terminals. We studied sprouting of optic fibers into an area denervated by local microinjection of beta-bungarotoxin (beta-BTX), a specific presynaptic neurotoxin with phospholipase A2 activity that destroys nerve terminals at the neuromuscular junction. After injection of 0.1 pmol of beta-BTX, the optic terminals fired spontaneously with decreasing amplitude and became silent within 1 to 2 h. Outside the injection zone, the retinotectal map was normal, so the silent zone was associated with a scotoma in the visual field. Horseradish peroxidase (HRP) staining of the entire optic nerve showed a denervated region at the injection site with beaded, degenerating fibers at its edge. Between 3 and 9 days later, optic units were recorded within the injection zone whose receptive fields lay just outside the scotoma in the visual field, indicating that intact surrounding terminals had sprouted into the area. These sprouts made functional connections, as indicated by field potential recordings and current source-density analysis. At this time, HRP staining also demonstrated retinal innervation within the injection zone. By 12 days, normal maps with no scotoma were recorded and HRP staining was normal at the injection site, indicating that the beta-BTX-damaged fibers had regenerated to reclaim their tectal sites. The results show that the retinotectal projection of goldfish is very dynamic, since intact optic fibers can sprout into adjacent vacant postsynaptic territory within 2 to 3 days, much faster than previously reported. In a final experiment, we showed that this sprouting is activity-dependent, since it could be prevented by blocking retinal activity with intraocular tetrodotoxin (TTX) during the first 2 days postinjection, even though TTX block of activity does not block regeneration in this system. One possible mechanism for this rapidly triggered sprouting is that arachidonic acid liberated by beta-BTX acts as a sprouting factor to attract surrounding healthy fibers into the denervated region but requires activity at the terminals to be effective.


Subject(s)
Nerve Regeneration/physiology , Optic Nerve/physiology , Animals , Bungarotoxins , Denervation , Goldfish , Horseradish Peroxidase , Nerve Regeneration/drug effects , Tectum Mesencephali , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...