Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Exp Med ; 184(4): 1425-33, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8879214

RESUMEN

In Alzheimer's disease (AD), affected neurons accumulate beta amyloid protein, components of which can induce mouse microglia to express the high-output isoform of nitric oxide synthase (NOS2) in vitro. Products of NOS2 can be neurotoxic. In mice, NOS2 is normally suppressed by transforming growth factor beta 1 (TGF-beta 1). Expression of TGF-beta 1 is decreased in brains from AD patients, a situation that might be permissive for accumulation of NOS2. Accordingly, we investigated the expression of NOS2 in patients with AD, using three monospecific antibodies: a previously described polyclonal and two new monoclonal antibodies. Neurofibrillary tangle-bearing neurons and neuropil threads contained NOS2 in brains from each of 11 AD patients ranging in age from 47 to 81 years. NOS2 was undetectable in brains from 6 control subjects aged 23-72 years, but was expressed in small amounts in 3 control subjects aged 77-87 years. Thus, human neurons can express NOS2 in vivo. The high-output pathway of NO production may contribute to pathogenesis in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Encéfalo/enzimología , Isoenzimas/aislamiento & purificación , Neuronas/enzimología , Óxido Nítrico Sintasa/aislamiento & purificación , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Anticuerpos Monoclonales , Especificidad de Anticuerpos , Encéfalo/patología , Inducción Enzimática , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Neuronas/patología
2.
J Cell Biol ; 147(1): 121-34, 1999 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-10508860

RESUMEN

Presenilin 1 (PS1) is the causative gene for an autosomal dominant familial Alzheimer's disease (AD) mapped to chromosome 14. Here we show that QM/Jun-interacting factor (Jif)-1, a negative regulator of c-Jun, is a candidate to mediate the function of PS1 in the cell. We screened for proteins that bind to PS1 from a human embryonic brain cDNA library using the two-hybrid method and isolated one clone encoding the QM/Jif-1 gene. The binding of QM/Jif-1 to full-length PS1 was confirmed in vitro by pull-down assay, and in vivo by immunoprecipitation assays with human samples, including AD brains. Immunoelectronmicroscopic analysis showed that QM/Jif-1 and PS1 are colocalized at the endoplasmic reticulum, and the nuclear matrix in human brain neurons. Chloramphenicol acetyltransferase assays in F9 cells showed that PS1 suppresses transactivation by c-Jun/c-Jun but not by c-Jun/c-Fos heterodimers, consistent with the reported function of QM/Jif-1. By monitoring fluorescent recombinant protein and by gel mobility shift assays, PS1 was shown to accelerate the translocation of QM from the cytoplasm to the nucleus and to thereby suppress the binding of c-Jun homodimer to 12-O-tetradecanoylphorbol-13- acetate (TPA)-responsive element (TRE). PS1 suppressed c-jun-associated apoptosis by retinoic acid in F9 embryonic carcinoma cells, whereas this suppression of apoptosis is attenuated by mutation in PS1. Collectively, the novel function of PS1 via QM/Jif-1 influences c-jun-mediated transcription and apoptosis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Ribosómicas , Adulto , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Animales , Apoptosis , Transporte Biológico , Encéfalo/citología , Encéfalo/embriología , Encéfalo/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/genética , Núcleo Celular/metabolismo , Dimerización , Femenino , Humanos , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Mutación , Presenilina-1 , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Proteína Ribosómica L10 , Activación Transcripcional , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Dedos de Zinc
3.
J Neurol Sci ; 276(1-2): 9-13, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18848708

RESUMEN

Mutations in the gene progranulin (PGRN) were recently identified as the cause of some forms of frontotemporal dementia with ubiquitin-positive intraneuronal inclusion pathology (FTLD-U). The DNA-binding protein, TDP-43, was determined to be a component of these ubiquitinated inclusions in FTLD-U and amyotrophic lateral sclerosis (ALS) with dementia (ALS-D). These findings raise many interesting questions as to the shared pathology and possible common pathologic process between ALS and FTLD-U. This study examines the immunoexpression of PGRN in ALS patients using immunohistochemical analysis of post-mortem tissue. Available brain and spinal cord sections of eight ALS patients, including one case with severe dementia, and eighteen control-aged brains were stained with anti-PGRN antibodies. We found increased staining for PGRN in motor tracts with vacuolar degeneration and glial cells in ALS sample spinal cord and brainstem sections compared to controls. Variable upper motor neuron staining and reactive glia were seen in ALS motor cortex samples. Frontal lobe and hippocampal sections showed no consistent differences from control tissues with the exception of the ALS-dementia case, which showed PGRN immunoexpression in non-motor cortical areas. These results describe a pattern of increased PGRN expression in areas of active degeneration in ALS. The meaning of this association is unclear, but may indicate a potential role for PGRN in the variable expression of motor and cognitive deficits in the ALS-FTD spectrum.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Anciano , Esclerosis Amiotrófica Lateral/complicaciones , Esclerosis Amiotrófica Lateral/patología , Encéfalo/metabolismo , Encéfalo/patología , Demencia/complicaciones , Demencia/metabolismo , Demencia/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Progranulinas , Médula Espinal/metabolismo
4.
J Neurol Sci ; 277(1-2): 164-6, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19013587

RESUMEN

Dystonia associated with neurodegenerative disease has minimal effective treatment options and can be devastating to a patient's ability to perform tasks of daily living. We present a case of a 55 year-old man who had progressive symptoms of an atypical asymmetric parkinsonian neurodegenerative disease. This patient presented with a dystonic left upper extremity that was refractory to treatment. In an attempt to treat worsening pain associated with the dystonia, he was given a five-day lidocaine infusion for associated pain and within 24 h had improvement in mobility of his dystonic extremity. Dystonia was measured by the Burke-Fahn-Marsden (BFM) dystonia rating and disability scales on hospital day five and at an eight week follow up visit. These scores were compared with scores derived from his previous pre-treatment neurologic examination. The BFM dystonia scale score was initially 16 and improved to 12 on both immediate post-treatment and eight-week follow-up. The BFM disability score improved from 16 to 6 post treatment and to 8 on follow-up appointment. Most importantly, the patient could feed and dress himself for the first time in several years. No adverse events of treatment were encountered. Treatment effect lasted three months with a slow return to baseline motor function. This case report raises interesting questions regarding the mechanism of dystonia in neurodegenerative disease and suggests the afferent sensory system as a potential target for therapeutics.


Asunto(s)
Anestésicos Locales/administración & dosificación , Distonía/tratamiento farmacológico , Trastornos Distónicos/tratamiento farmacológico , Lidocaína/administración & dosificación , Enfermedades Neurodegenerativas/complicaciones , Distonía/etiología , Trastornos Distónicos/etiología , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Neurodegenerativas/diagnóstico por imagen , Tomografía Computarizada por Rayos X
5.
Prog Neurobiol ; 54(1): 71-85, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9460794

RESUMEN

Transforming growth factors-betas (TGF-betas), a family of multifunctional peptide growth factors, affect cells of the central nervous system (CNS). The three mammalian TGF-beta isoforms, TGF-betas 1, 2 and 3, are expressed in adult human brain. Since neuronal degeneration is a defining feature of CNS degenerative diseases, TGF-beta may be important because it can influence neuronal survival. In vitro TGF-beta promotes survival of rat spinal cord motoneurons and dopaminergic neurons. In addition to direct effects on neuronal survival, TGF-beta treatment of cultured astrocytes induces a reactive phenotype. Thus, TGF-beta may also normalize the extracellular matrix environment in degenerative diseases. The expression of TGF-betas change in response to neuronal injury. TGF-beta 1 expression increases in astrocytes and microglia in animal models of cerebral ischemia, while TGF-beta 2 expression increases in activated astroglial cells in human neurodegenerative diseases. TGF-betas protect neurons from a variety of insults. TGF-beta maintains survival of chick telencephalic neurons made hypoxic by treatment with cyanide and decreases the area of infarction when administered in animal models of cerebral ischemia. In vitro TGF-beta protects neurons from damage induced by treatment with beta-amyloid peptide, FeSO4 (induces production of reactive oxygen species), Ca2+ ionophores, glutamate, glutamate receptor agonists and MPTP (toxic for dopaminergic neurons). TGF-beta maintains mitochondrial potential and Ca2+ homeostasis and inhibits apoptosis in neurons. TGF-beta does not prevent neuronal degeneration in a rat model of Parkinson's disease and has yet to be tested in newly developed transgenic mouse models of Alzheimer's disease. TGF-beta is a potent neuroprotective agent which may affect the pathogenesis of neurodegenerative diseases of the CNS.


Asunto(s)
Enfermedades Neurodegenerativas/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedades Neurodegenerativas/patología , Factor de Crecimiento Transformador beta/química , Factor de Crecimiento Transformador beta/metabolismo
6.
Curr Mol Med ; 1(6): 733-7, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11899259

RESUMEN

Alzheimer's disease (AD) is a common neurodegenerative disease that affects cognitive function in the elderly. Large extracellular beta-amyloid (Abeta) plaques and tau-containing intraneuronal neurofibrillary tangles characterize AD from a histopathologic perspective. However, the severity of dementia in AD is more closely related to the degree of the associated neuronal and synaptic loss. It is not known how neurons die and synapses are lost in AD; the current review summarizes what is known about this issue. Most evidence indicates that amyloid precursor protein (APP) processing is central to the AD process. The Abeta in plaques is a metabolite of the APP that forms when an alternative (beta-secretase and then gamma-secretase) enzymatic pathway is utilized for processing. Mutations of the APP gene lead to AD by influencing APP metabolism. One leading theory is that the Abeta in plaques leads to AD because Abeta is directly toxic to the adjacent neurons. Other theories advance the notion that neuronal death is triggered by intracellular events that occur during APP processing or by extraneuronal preplaque Abeta oligomers. Some investigators speculate that in many cases there is a more general disorder of protein processing in neurons that leads to cell death. In the later models, Abeta plaques are a byproduct of the disease process, rather than the direct cause of neuronal death. A direct correlation between Abeta plaque burden and neuronal (or synaptic) loss should occur in AD if Abeta plaques cause AD through a direct toxic effect. However, histopathologic studies indicate that the correlation between Abeta plaque burden and neuronal (or synaptic) loss is poor. We conclude that APP processing and Abeta formation is important to the AD process, but that neuronal alterations that underlie symptoms of AD are not due exclusively to a direct toxic effect of the Abeta deposits that occur in plaques. A more general problem with protein processing, damage due to the neuron from accumulation of intraneuronal Abeta or extracellular, preplaque Abeta may also be important as underlying factors in the dementia of AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/fisiología , Neuronas/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Muerte Celular/fisiología , Humanos , Modelos Neurológicos , Procesamiento Proteico-Postraduccional
7.
J Neuropathol Exp Neurol ; 54(3): 297-303, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7745428

RESUMEN

Cell cycle-associated nuclear proteins may have more specialized functions in the adult nervous system in addition to those directly associated with cell proliferation, as suggested by a recent study showing that neurofibrillary tangles (NFT) and dystrophic neurites in Alzheimer's disease (AD) are immunoreactive for the proliferation-associated antigen p105. To further investigate this hypothesis, we studied the expression of another proliferation-associated antigen, Ki-67, in the brains of patients with AD and other neurodegenerative disorders. Formalin-fixed, paraffin-embedded sections from autopsy cases of AD, Down's syndrome with dementia and AD pathology (DS/AD), Pick's disease (PiD), progressive supranuclear palsy (PSP), Lewy body disease (LBD), Parkinson's disease (PD), corticobasal degeneration (CBD), and young and aged normal brains, and from two surgically resected gangliogliomas were immunostained using antibodies to Ki-67 (MIB-1 clone equivalent) and tau (tau). Ki-67 staining was performed following antigen retrieval by microwave heating. Ki-67 labeled NFT that were observed in the AD, DS/AD, PiD, PSP, LBD, and PD cases, one aged normal brain, and one ganglioglioma. Ki-67 generally labeled fewer NFT compared to tau. Pick bodies, ballooned neurons (Pick cells) in CBD and PiD, and nigral corticobasal inclusions in CBD were immunoreactive for tau but not Ki-67. Neither antibody labeled cortical or subcortical Lewy bodies. Our findings suggest that Ki-67 may be involved in the pathogenesis of neurofibrillary degeneration in AD, other neurodegenerative disorders, normal aging, and neoplasms such as ganglioglioma. We postulate a possible role for Ki-67 in the production of the abnormally phosphorylated tau protein that leads to the formation of paired helical filaments within susceptible neurons.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Proteínas de Neoplasias/análisis , Proteínas Nucleares/análisis , Adulto , Anciano , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Demencia/inmunología , Demencia/patología , Síndrome de Down/complicaciones , Síndrome de Down/inmunología , Femenino , Humanos , Antígeno Ki-67 , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/química , Enfermedad de Parkinson/inmunología , Parálisis Supranuclear Progresiva/inmunología
8.
J Neuropathol Exp Neurol ; 61(11): 935-46, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12430710

RESUMEN

A working group supported by the Office of Rare Diseases of the National Institutes of Health formulated neuropathologic criteria for corticobasal degeneration (CBD) that were subsequently validated by an independent group of neuropathologists. The criteria do not require a specific clinical phenotype, since CBD can have diverse clinical presentations, such as progressive asymmetrical rigidity and apraxia, progressive aphasia, or frontal lobe dementia. Cortical atrophy, ballooned neurons, and degeneration of the substantia nigra have been emphasized in previous descriptions and are present in CBD, but the present criteria emphasize tau-immunoreactive lesions in neurons, glia, and cell processes in the neuropathologic diagnosis of CBD. The minimal pathologic features for CBD are cortical and striatal tau-positive neuronal and glial lesions, especially astrocytic plaques and thread-like lesions in both white matter and gray matter, along with neuronal loss in focal cortical regions and in the substantia nigra. The methods required to make this diagnosis include histologic stains to assess neuronal loss, spongiosis and ballooned neurons, and a method to detect tau-positive neuronal and glial lesions. Use of either the Gallyas silver staining method or immunostains with sensitive tau antibodies is acceptable. In cases where ballooned neurons are sparse or difficult to detect, immunostaining for phospho-neurofilament or alpha-B-crystallin may prove helpful. Methods to assess Alzheimer-type pathology and Lewy body pathology are necessary to rule out other causes of dementia and Parkinsonism. Using these criteria provides good differentiation of CBD from other tauopathies, except frontotemporal dementia and Parkinsonism linked to chromosome 17, where additional clinical or molecular genetic information is required to make an accurate diagnosis.


Asunto(s)
Encéfalo/patología , Enfermedades Neurodegenerativas/patología , Ovillos Neurofibrilares/patología , Neuroglía/patología , Neuronas/patología , Atrofia/patología , Atrofia/fisiopatología , Encéfalo/fisiopatología , Humanos , Cuerpos de Inclusión/patología , Enfermedades Neurodegenerativas/fisiopatología , Tinción con Nitrato de Plata
9.
Neurobiol Aging ; 11(5): 551-4, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2234286

RESUMEN

The histopathology of the indusium griseum (IG), a displaced hippocampal anlage, was studied in five patients with Alzheimer's disease (AD) and five controls. In the AD group, the IG had occasional neurons with granulovacuolar change (GVD) and rare Hirano bodies (HB), but no senile plaques (SP), neurofibrillary tangles (NFT), or neurons staining for phosphorylated neurofilament antigen. There was a slight but not statistically significant diminution of neurons within the IG. In all AD cases, the hippocampus showed abundant AD-associated histopathology. In the control cases, only rare neurons with GVD were present in the IG. These findings indicate that although single neurons within the IG may show some of the cytologic changes seen in the hippocampal neurons in normal aging and AD, IG neurons do not express the full range and severity of histopathologic abnormalities characteristic of AD. This suggests that factors other than selective vulnerability of neurons of hippocampal origin might be operating to induce the neuropathologic picture of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Hipocampo/patología , Neurofibrillas/patología , Neuronas/patología , Anciano , Anciano de 80 o más Años , Femenino , Hipocampo/anatomía & histología , Humanos , Inmunohistoquímica , Masculino
10.
Neurobiol Aging ; 19(6): 527-33, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10192211

RESUMEN

The transforming growth factor-betas (TGF-betas) influence cell survival, and TGF-beta2 shows increased immunoexpression in neurofibrillary tangle-bearing neurons and reactive glia in Alzheimer's disease (AD) and progressive supranuclear palsy (PSP). We compared immunohistochemical expression of TGF-beta type I (RI) and type II (RII) receptors in eight patients with AD, eight controls and three cases of progressive supranuclear palsy. Mild intraneuronal immunoreactivity for the RI receptor was observed in all cases. Intraneuronal TGF-beta RII receptor immunoexpression was more common in all groups, and its frequency did not differ between groups. We observed increased immunoreactivity for both RI and RII subtypes in reactive glia in the AD frontal cortex (RI: U = 0.5, p = 0.002; and RII: U = 9.000, p = 0.006) and parahippocampal gyrus (RI: U = 9.500, p = 0.013; RII: U = 14.5, p = 0.05) compared to control cases. We conclude that TGF-beta RI and II immunoreactivity is increased in reactive glia in AD and progressive supranuclear palsy, and RI immunoreactivity may occasionally be increased in neurons in cases with advanced AD.


Asunto(s)
Receptores de Activinas Tipo I , Enfermedad de Alzheimer/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Parálisis Supranuclear Progresiva/metabolismo , Anciano , Envejecimiento/metabolismo , Enfermedad de Alzheimer/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Parálisis Supranuclear Progresiva/patología
11.
Neurobiol Aging ; 13(3): 405-11, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1625770

RESUMEN

The hippocampal perforant pathway originates in the entorhinal cortex (ERC) and terminates in the outer molecular layer of the dentate gyrus (DG). To compare the effects of normal aging and Alzheimer's disease (AD) on the elements of the perforant pathway, we compared relative perikaryal numbers (determined by counting cell bodies and estimating volumes) in layer II of the ERC with synaptic quantities (estimated from immunoreactivity for the synaptic terminal protein synapsin I and DG volume) in the molecular layer of the DG. The brains of 5 young and 9 elderly cognitively normal individuals, and of 9 AD patients were studied. In normal aging we found a significant age-related decline in perikaryal numbers in the ERC without demonstrable synaptic loss in the DG. In AD there was marked and equivalent, (or proportional) reduction in both ERC perikaryal numbers and DG synapses. These data suggest that in normal aging remaining neurons may continue to support a full array of synapses, perhaps due to mechanisms such as axonal sprouting, synaptic enlargement, or synaptic ingrowth. In AD, however, the accelerated neuronal loss may overwhelm such compensatory mechanisms or alternatively, independent synaptic and perikaryal losses may occur.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/patología , Vías Nerviosas/patología , Sinapsis/patología , Adulto , Anciano , Anciano de 80 o más Años , Preescolar , Hipocampo/patología , Humanos , Procesamiento de Imagen Asistido por Computador , Persona de Mediana Edad , Lóbulo Temporal/patología
12.
Arch Neurol ; 52(11): 1074-9, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7487559

RESUMEN

BACKGROUND: Published studies now show a clear association between Alzheimer's disease (AD) and the apolipoprotein E epsilon 4 allele (APOE* epsilon 4). The clinical value of this information to estimate a healthy individual's lifetime risk of AD has not been well delineated. Physicians dealing with AD may not know either the lifetime risk of developing AD or the effect of the APOE genotype on this risk. Because the lifetime risk of AD depends in part on life expectancy, and available figures on APOE are not population based, a computation is necessary to derive risk estimates useful to physicians. OBJECTIVES: To estimate the lifetime risk of AD and the effect of APOE genotype information on that risk and to assess the knowledge of these risks among physicians who manage patients with dementia. DESIGN: Estimation of risk of AD and survey of physician awareness. The lifetime risk of developing AD without APOE genotype information was first computed for 65-year-olds from existing epidemiologic studies of age-related AD incidence and an actuarial life-table analysis. Using this computed a priori risk of AD and published studies of APOE genotypes in individuals with and without AD, we used a Bayesian analysis to determine the risk of developing AD, with and without an APOE* epsilon 4 allele, for unaffected 65-year-olds. To assess physician knowledge of the lifetime risk of AD and the effect of APOE genotyping on the risk, 50 neurologists, internists, geriatricians, geriatric psychiatrists, and family physicians who manage patients with dementia were randomly selected to participate in a questionnaire-driven telephone survey. RESULTS: In a person with no family history of AD, the epidemiologic/actuarial lifetime risk of AD is approximately 15%. Based on a Bayesian calculation and published APOE data, the lifetime risk of AD is 29% for individuals with one APOE* epsilon 4 allele and it is 9% if no APOE* epsilon 4 allele is present. Physician awareness survey results were as follows: 42% of physicians correctly estimated the approximate lifetime risk of AD; of these, only one third were moderately sure of their response. Only three physicians correctly estimated the change in risk given the APOE* epsilon 4 genotype; only one of these was at least moderately sure. CONCLUSIONS: Determining the APOE* epsilon 4 status of healthy adults with no family history of AD approximately doubles (for the epsilon 4 allele) or reduces by 40% (for the non-epsilon 4 allele) the uninformed lifetime risk of developing AD. Even with an APOE* epsilon 4 allele, the lifetime risk remains below 30%. Most physicians managing patients with AD do not know the lifetime risk of AD, and very few know how APOE* epsilon 4 status modifies the risk. These clinically relevant risk figures should be more widely disseminated among physicians.


Asunto(s)
Alelos , Enfermedad de Alzheimer/genética , Apolipoproteínas E/genética , Análisis Actuarial , Adulto , Anciano , Enfermedad de Alzheimer/epidemiología , Apolipoproteína E4 , Teorema de Bayes , Humanos , Factores de Riesgo
13.
Arch Neurol ; 50(10): 1088-92, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8215968

RESUMEN

OBJECTIVE: To determine the relationship between vascular beta-amyloid (beta A4) and senile plaques (SPs) and neurofibrillary tangles (NFTs). DESIGN: We counted vascular amyloid deposition with SP and NFT density in the medial temporal lobe (CA1 plus the subiculum) and the cerebellum. PATIENTS: The brains of seven patients with Alzheimer's disease and of three age-matched nondemented control subjects were studied. RESULTS: In Alzheimer's disease, the density of beta A4-laden blood vessels was significantly higher in the cerebellum than in CA1 plus the subiculum. Conversely, the densities of SPs and NFTs were much greater in the CA1 plus the subiculum than in the cerebellum. CONCLUSIONS: This study indicates that local vascular beta A4 deposition is not directly correlated with SP and NFT densities. Deposition of beta A4 in blood vessel walls may not be instrumental in the formation of SPs and/or NFTs in the brain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/análisis , Química Encefálica , Encéfalo/irrigación sanguínea , Ovillos Neurofibrilares/patología , Anciano , Anciano de 80 o más Años , Cerebelo/química , Cerebelo/patología , Hipocampo/química , Hipocampo/patología , Humanos , Persona de Mediana Edad , Lóbulo Temporal/química , Lóbulo Temporal/patología
14.
Arch Neurol ; 58(11): 1817-20, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11708989

RESUMEN

BACKGROUND: Alpha-synuclein is a major component of Lewy bodies (LBs) in Parkinson disease and dementia with LBs and of glial cytoplasmic inclusions in multiple system atrophy. However, epitope mapping for alpha-synuclein is distinctive in different neurodegenerative diseases. The reasons for this are poorly understood but may reflect fundamental differences in disease mechanisms. OBJECTIVE: To investigate the alpha-synuclein epitope mapping properties of LBs in familial Alzheimer disease. DESIGN AND SETTING: We compared LBs in familial Alzheimer disease with those in synucleinopathies by probing 6 brains of persons with familial Alzheimer disease using a panel of antibodies to epitopes spanning the alpha-synuclein protein. Results were compared with data from brains of persons with Parkinson disease, dementia with LBs, and multiple system atrophy. RESULTS: The brains of persons with familial Alzheimer disease showed consistent staining of LBs with all antibodies, similar to Parkinson disease and dementia with LBs but different from alpha-synuclein aggregates that occurred in multiple system atrophy. CONCLUSIONS: These data suggest that the epitope profiles of alpha-synuclein in LBs are similar, regardless of whether the biological trigger is related to synuclein or a different genetic pathway. These findings support the hypothesis that the mechanism of alpha-synuclein aggregation is the same within cell types but distinctive between cell types.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Mapeo Epitopo , Cuerpos de Lewy/química , Proteínas del Tejido Nervioso/análisis , Anciano , Enfermedad de Alzheimer/genética , Anticuerpos Monoclonales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Enfermedad por Cuerpos de Lewy/patología , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/patología , Sinucleínas , alfa-Sinucleína
15.
Arch Neurol ; 56(9): 1111-8, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10488812

RESUMEN

BACKGROUND: Alterations in the metabolism of the amyloid precursor protein and the formation of beta-amyloid (Abeta) plaques are associated with neuronal death in Alzheimer disease (AD). The plaque subtype Abeta(x-42) occurs as an early event, with Abeta(x-40) plaques forming at a later stage. In dementia with Lewy bodies (DLB), an increase in the amount of cortical Abeta occurs without severe cortical neuronal losses. OBJECTIVE: To advance our understanding of the natural history of Abeta in neurodegenerative diseases. DESIGN: We evaluated the expression of Abeta(x-40) and Abeta(x-42) in DLB using monoclonal antibodies and immunohistochemical techniques in 5 brain regions. The data were compared with those elicited with normal aging and from patients with AD. SETTING AND PATIENTS: A postmortem study involving 19 patients with DLB without concurrent neuritic degeneration, 10 patients with AD, and 17 aged persons without dementia for control subjects. RESULTS: The Abeta plaques were more numerous in patients with DLB than in controls in most brain regions, although the Abeta(x-42) plaque subtype was predominant in both conditions. Overall, Abeta(x-42) plaque density was similar in patients with DLB and those with AD, but Abeta(x-40) plaques were more numerous in persons with AD than in those with DLB. The ratio of Abeta(x-40) to Abeta(x-42) plaques was significantly reduced in persons with DLB compared with patients with AD. CONCLUSIONS: The Abeta plaques were more numerous in patients with DLB than persons with normal aging, but the plaque subtypes were similar. The relative proportion of the 2 Abeta plaque subtypes in DLB is distinguishable from that in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/análisis , Demencia/metabolismo , Cuerpos de Lewy/química , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Anticuerpos Monoclonales , Química Encefálica , Estudios de Casos y Controles , Demencia/patología , Femenino , Lóbulo Frontal/química , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad
16.
Neurology ; 41(6): 882-6, 1991 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2046934

RESUMEN

We describe the clinical, radiologic, neuropsychological, and neuropathologic features of a 69-year-old man with a 3-year history of progressive transcortical expressive aphasia. Neuropsychological testing showed progressive dysfunction of expressive language. Neuropathologic examination demonstrated focal cortical degeneration involving the left superior frontal gyrus, with swollen achromasic neurons and no evidence of Alzheimer's disease, Pick's disease, Creutzfeldt-Jakob disease, Lewybody disease, or other dementing disorders. This case adds to the known heterogeneity of the underlying pathology of patients with primary progressive aphasia.


Asunto(s)
Afasia/patología , Encefalopatías/patología , Anciano , Afasia/complicaciones , Afasia/psicología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encefalopatías/complicaciones , Encefalopatías/psicología , Humanos , Masculino , Pruebas Neuropsicológicas , Tomografía Computarizada por Rayos X
17.
Neurology ; 45(8): 1561-9, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7543987

RESUMEN

We compared immunohistochemical expression of the transforming growth factor-betas (TGF-beta 1, TGF-beta 2, and TGF-beta 3) using brain tissue from patients with nondominantly inherited Alzheimer's disease (NDAD) (n = 9), autosomal dominantly inherited Alzheimer's disease with linkage to 14q24.3 (FAD-14) (n = 4), and cognitively normal controls (n = 10) to determine whether their pathologic changes are associated with an altered distribution of the TGF-betas. We found increased expression of TGF-beta 2 in large, tangle-bearing neurons with widespread staining of glia in NDAD and FAD-14 patients compared with control cases. This result was confirmed with sandwich ELISA assays of brain tissue, which showed TGF-beta 2 levels in AD and NDAD to average 3.2 times the average level of control cases. Despite proximity of TGF-beta 1 and TGF-beta 3 to the sites of susceptibility loci on chromosomes 19 and 14, we did not find that TGF-beta 1 and TGF-beta 3 were selectively altered in any AD subtypes. However, selective induction of TGF-beta 2 may occur in NDAD and FAD-14.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Cromosomas Humanos Par 14 , Demencia/metabolismo , Demencia/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Genes Dominantes , Ligamiento Genético , Humanos , Inmunohistoquímica/métodos , Masculino , Persona de Mediana Edad , Valores de Referencia , Coloración y Etiquetado
18.
Neurology ; 54(1): 100-4, 2000 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-10636133

RESUMEN

OBJECTIVE: To assess AMY expression in familial AD (FAD). BACKGROUND: The discovery of nonbeta-amyloid (Abeta), plaque-like deposits composed of a 100-kd protein (AMY) in sporadic AD (SAD) brains prompted us to determine whether these plaques (AMY plaques) also occur in AD due to mutations of the presenilin-1 (PS-1), presenilin-2 (PS-2), or the amyloid precursor protein (APP) genes. METHODS: We used immunohistochemistry and confocal laser scanning microscopy to probe the brains of 22 patients with FAD (13 with PS-1, 5 with PS-2, and 4 with APP mutations) and 14 patients with SAD. RESULTS: AMY plaques were present in all SAD and FAD brains, including an FAD/PS-1 brain from an individual with preclinical disease. The morphology of AMY plaques in SAD and FAD brains was indistinguishable, but they differed from Abeta deposits because AMY plaques lacked an immunoreactive core. AMY plaques sometimes colocalized with Abeta(x-42) deposits, but they did not colocalize with Abeta(x-40) plaque cores in either SAD or FAD brains. The percent of cortical area occupied by AMY was greater in FAD than in SAD brains (mean percent area = 9.8% and 5.9%, t = 2.487, p = 0.018). In particular, APP and PS-1 cases had more AMY deposition than PS-2 or SAD cases (12.9%, 10.5%, 6.2% in APP, PS-1, and PS-2 AD). CONCLUSIONS: AMY plaques are consistently present in familial AD due to presenilin-1 (PS-1), PS-2, and amyloid precursor protein mutations, and they can begin to accumulate before the emergence of dementia.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Placa Amiloide/patología , Anciano , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Encéfalo/metabolismo , Humanos , Inmunohistoquímica , Masculino , Proteínas de la Membrana/genética , Microscopía Confocal , Persona de Mediana Edad , Mutación , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1 , Presenilina-2
19.
Neurology ; 56(11 Suppl 4): S31-4, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11402148

RESUMEN

The authors describe a 49-year-old woman (R.K.) who presented with one year of progressive frontal lobe dysfunction, including signs of expressive aphasia. Signs of parkinsonism were absent until late in the clinical course. Neuropsychologic testing and neuroimaging studies are described. The patient died at age 55, after 7 years of symptoms. Family history was remarkable for a mother who died at the age of 45, after experiencing 7 years of progressive aphasia. R.K.'s brain showed asymmetric frontotemporal atrophy, which was more severe on the left side. Histopathologic analysis was remarkable for numerous tau-positive neurons with some classic-appearing Pick bodies and many ballooned neurons. Tau-positive glial cells were also present. The authors suggest that the abnormal tau aggregates are related to the symptoms experienced by affected members of this family.


Asunto(s)
Encéfalo/patología , Demencia/patología , Neuroglía/patología , Neuronas/patología , Proteínas tau/metabolismo , Afasia/genética , Demencia/genética , Femenino , Lateralidad Funcional , Humanos , Persona de Mediana Edad , Pruebas Neuropsicológicas , Linaje
20.
Neurology ; 48(2): 515-9, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9040748

RESUMEN

To determine whether apolipoprotein E epsilon 2 (APOE-epsilon 2) affects neuropathology in aging and Alzheimer's disease (AD), we compared beta-amyloid plaque (A beta P) and neurofibrillary tangle densities, neuropil thread formation, and amyloid angiopathy in five APOE-epsilon 2/3 AD patients, five APOE-epsilon 3/3 AD patients, five APOE-epsilon control patients, and five APOE-epsilon 3/3 control patients. We examined the (frontal and parietal) neocortex, hippocampus, entorhinal cortex, and cerebellum and found A beta P densities to be lower (t = 3.121, p = 0.011) in the cortex of APOE-epsilon 2/3 AD patients than in APOE-epsilon 3/3 AD patients. Amyloid angiopathy was also less in APOE-epsilon 2/3 patients than in APOE-3/3 patients (U = 4.500, p = 0.027). Control APOE-epsilon 2/3 brains had little AD-related pathology; even our 102-year-old control case showed few A beta Ps compared with the elderly APOE-epsilon 3/3 cases. The APOE-epsilon 2/3 genotype may influence pathologic phenotype in some aged normal and AD populations.


Asunto(s)
Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Ovillos Neurofibrilares/patología , Anciano , Alelos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E2 , Apolipoproteína E3 , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA