Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
AJNR Am J Neuroradiol ; 40(6): 938-945, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31147354

RESUMEN

BACKGROUND AND PURPOSE: Accurate automated infarct segmentation is needed for acute ischemic stroke studies relying on infarct volumes as an imaging phenotype or biomarker that require large numbers of subjects. This study investigated whether an ensemble of convolutional neural networks trained on multiparametric DWI maps outperforms single networks trained on solo DWI parametric maps. MATERIALS AND METHODS: Convolutional neural networks were trained on combinations of DWI, ADC, and low b-value-weighted images from 116 subjects. The performances of the networks (measured by the Dice score, sensitivity, and precision) were compared with one another and with ensembles of 5 networks. To assess the generalizability of the approach, we applied the best-performing model to an independent Evaluation Cohort of 151 subjects. Agreement between manual and automated segmentations for identifying patients with large lesion volumes was calculated across multiple thresholds (21, 31, 51, and 70 cm3). RESULTS: An ensemble of convolutional neural networks trained on DWI, ADC, and low b-value-weighted images produced the most accurate acute infarct segmentation over individual networks (P < .001). Automated volumes correlated with manually measured volumes (Spearman ρ = 0.91, P < .001) for the independent cohort. For the task of identifying patients with large lesion volumes, agreement between manual outlines and automated outlines was high (Cohen κ, 0.86-0.90; P < .001). CONCLUSIONS: Acute infarcts are more accurately segmented using ensembles of convolutional neural networks trained with multiparametric maps than by using a single model trained with a solo map. Automated lesion segmentation has high agreement with manual techniques for identifying patients with large lesion volumes.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador/métodos , Redes Neurales de la Computación , Neuroimagen/métodos , Anciano , Imagen de Difusión por Resonancia Magnética/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Accidente Cerebrovascular/diagnóstico por imagen
2.
Neurology ; 72(14): 1230-5, 2009 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-19349602

RESUMEN

BACKGROUND: Neuropathologic studies suggest an association between cerebral amyloid angiopathy (CAA) and small ischemic infarctions as well as hemorrhages. We examined the prevalence and associated risk factors for infarcts detected by diffusion-weighted imaging (DWI). METHODS: We performed retrospective analysis of MR images from 78 subjects with a diagnosis of probable CAA and a similar aged group of 55 subjects with Alzheimer disease or mild cognitive impairment (AD/MCI) for comparison. DWI and apparent diffusion coefficient (ADC) maps were inspected for acute or subacute infarcts. We also examined the association between DWI lesions and demographic variables, conventional vascular risk factors, and radiographic markers of CAA severity such as number of hemorrhages on gradient-echo MRI and volume of T2-hyperintense white matter lesions. RESULTS: Twelve of 78 subjects with CAA (15%) had a total of 17 DWI-hyperintense lesions consistent with subacute cerebral infarctions vs 0 of 55 subjects with AD/MCI (p = 0.001). The DWI lesions were located primarily in cortex and subcortical white matter. CAA subjects with DWI lesions had a higher median number of total hemorrhages (22 vs 4, p = 0.025) and no difference in white matter hyperintensity volume or conventional vascular risk factors compared to subjects with CAA without lesions. CONCLUSIONS: MRI evidence of small subacute infarcts is present in a substantial proportion of living patients with advanced cerebral amyloid angiopathy (CAA). The presence of these lesions is associated with a higher burden of hemorrhages, but not with conventional vascular risk factors. This suggests that advanced CAA predisposes to ischemic infarction as well as intracerebral hemorrhage.


Asunto(s)
Angiopatía Amiloide Cerebral/complicaciones , Hemorragia Cerebral/etiología , Infarto Cerebral/etiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Encéfalo/patología , Angiopatía Amiloide Cerebral/patología , Hemorragia Cerebral/patología , Infarto Cerebral/patología , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Estudios Transversales , Recolección de Datos , Imagen de Difusión por Resonancia Magnética , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Estudios Longitudinales , Imagen por Resonancia Magnética , Masculino , Estudios Retrospectivos
3.
J Biol Chem ; 276(43): 40288-92, 2001 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-11544248

RESUMEN

The beta-amyloid precursor protein (APP) is a ubiquitous receptor-like molecule without a known function. However, the recent recognition that APP and Notch undergo highly similar proteolytic processing has suggested a potential signaling function for APP. After ligand binding, Notch is cleaved by the ADAM-17 metalloprotease followed by an intramembrane cleavage mediated by gamma-secretase. The gamma-secretase cut releases the Notch intracellular domain (NICD), which enters the nucleus and modulates transcription. Because APP is processed similarly by ADAM-17 and gamma-secretase, we reasoned that the APP intracellular domain (AICD) has a role analogous to the NICD. We therefore generated a plasmid encoding the AICD sequence and studied the subcellular localization of the expressed protein (C60). Our results demonstrate that the cytoplasmic domain of APP is a highly labile fragment that is stabilized by forming complexes with Fe65 and can then enter the nucleus in neurons and non-neural cells. These findings strongly support the hypothesis that APP signals in the nucleus in a manner analogous to the function of Notch.


Asunto(s)
Transporte Activo de Núcleo Celular , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Drosophila , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas ADAM , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Proteína de Unión a CREB , Endopeptidasas/metabolismo , Semivida , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Receptores Notch , Proteínas Represoras/metabolismo , Transducción de Señal , Transactivadores/metabolismo
4.
Neurobiol Dis ; 7(6 Pt B): 673-81, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11114265

RESUMEN

To investigate the mechanism of regulation of Ass production by familial Alzheimer's disease (FAD)-linked presenilin 1 (PS1), we used a cell-free system that allows de novo Ass generation to examine whether PS1 participates directly in the gamma-secretase reaction. Optimal Ass generation in vitro was achieved at mildly acidic pH and could be inhibited by the aspartyl protease inhibitor pepstatin A, consistent with the suggestion that gamma-secretase is an aspartyl protease. Dominant negative mutations of the critical transmembrane aspartates in PS1 or full deletion of PS1 did not alter the maturation of APP in the secretory pathway. Instead, PS1 had a direct effect on the inhibition of Ass production by a designed peptidomimetic inhibitor: the inhibition was significantly less effective in cells expressing FAD-causing mutations in either APP or PS1 than in cells expressing the wild-type proteins. Taken together, these findings suggest that PS1 participates physically in a complex with APP during the gamma-secretase cleavage event.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Células CHO , Fraccionamiento Celular , Sistema Libre de Células/metabolismo , Cricetinae , Endopeptidasas/efectos de los fármacos , Genes Dominantes , Aparato de Golgi/metabolismo , Concentración de Iones de Hidrógeno , Sustancias Macromoleculares , Proteínas de la Membrana/genética , Microsomas/metabolismo , Mutación Missense , Pepstatinas/farmacología , Presenilina-1 , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional
5.
Proc Natl Acad Sci U S A ; 97(16): 9299-304, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10922078

RESUMEN

An unusual intramembranous cleavage of the beta-amyloid precursor protein (APP) by gamma-secretase is the final step in the generation of amyloid beta-peptide (Abeta). Two conserved aspartates in transmembrane (TM) domains 6 and 7 of presenilin (PS) 1 are required for Abeta production by gamma-secretase. Here we report that the APP C-terminal fragments, C83 and C99, which are the direct substrates of gamma-secretase, can be coimmunoprecipitated with both PS1 and PS2. PS/C83 complexes were detected in cells expressing endogenous levels of PS. The complexes accumulate when gamma-secretase is inactivated either pharmacologically or by mutating the PS aspartates. PS1/C83 and PS1/C99 complexes were detected in Golgi-rich and trans-Golgi network-rich vesicle fractions. In contrast, complexes of PS1 with APP holoprotein, which is not the immediate substrate of gamma-secretase, occurred earlier in endoplasmic reticulum-rich vesicles. The major portion of intracellular Abeta at steady state was found in the same Golgi/trans-Golgi network-rich vesicles, and Abeta levels in these fractions were markedly reduced when either PS1 TM aspartate was mutated to alanine. Furthermore, de novo generation of Abeta in a cell-free microsomal reaction occurred specifically in these same vesicle fractions and was markedly inhibited by mutating either TM aspartate. Thus, PSs are complexed with the gamma-secretase substrates C83 and C99 in the subcellular locations where Abeta is generated, indicating that PSs are directly involved in the pathogenically critical intramembranous proteolysis of APP.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/química , Animales , Ácido Aspártico Endopeptidasas , Sitios de Unión , Células CHO , Cricetinae , Endopeptidasas/efectos de los fármacos , Aparato de Golgi/metabolismo , Humanos , Hidrólisis , Presenilina-1
6.
J Neurochem ; 75(2): 583-93, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10899933

RESUMEN

It has been hypothesized that a presenilin 1 (PS1)-related enzymatic activity is responsible for proteolytic cleavage of the C-terminal intracellular protein of Notch1, in addition to its role in beta-amyloid protein (Abeta) formation from the amyloid precursor protein (APP). We developed an assay to monitor ligand-induced Notch1 proteolysis and nuclear translocation in individual cells : Treatment of full-length Notch1-enhanced green fluorescent protein-transfected Chinese hamster ovary (CHO) cells with a soluble preclustered form of the physiologic ligand Delta leads to rapid accumulation of the C terminus of Notch1 in the nucleus and to transcriptional activation of a C-promoter binding factor 1 (CBF1) reporter construct. Nuclear translocation was blocked by cotransfection with Notch's physiologic inhibitor Numb. Using this assay, we now confirm and extend the observation that PS1 is involved in Notch1 nuclear translocation and signaling in mammalian cells. We demonstrate that the D257A and the D385A PS1 mutations, which had been shown previously to block APP gamma-secretase activity, also prevent Notch1 cleavage and translocation to the nucleus but do not alter Notch1 trafficking to the cell surface. We also show that two APP gamma-secretase inhibitors block Notch1 nuclear translocation with an IC(50) similar to that reported for APP gamma-secretase. Notch1 signaling, assessed by measuring the activity of CBF1, a downstream transcription factor, was impaired but not abolished by the PS1 aspartate mutations or gamma-secretase inhibitors. Our results support the hypotheses that (a) PS1-dependent APP gamma-secretase-like enzymatic activity is critical for both APP and Notch processing and (b) the Notch1 signaling pathway remains partially activated even when Notch1 proteolytic processing and nuclear translocation are markedly inhibited. The latter is an important finding from the perspective of therapeutic treatment of Alzheimer's disease by targeting gamma-secretase processing of APP to reduce Abeta production.


Asunto(s)
Ácido Aspártico , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares , Receptores de Superficie Celular , Factores de Transcripción , Sustitución de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas , Células CHO , Cricetinae , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Luciferasas/genética , Proteínas Luminiscentes/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mutación Puntual , Presenilina-1 , Presenilina-2 , Inhibidores de Proteasas/farmacología , Receptor Notch1 , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/metabolismo , Activación Transcripcional , Transfección
7.
Nat Cell Biol ; 2(7): 428-34, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10878808

RESUMEN

The beta-amyloid precursor protein (beta-APP), which is involved in the pathogenesis of Alzheimer's disease, and the Notch receptor, which is responsible for critical signalling events during development, both undergo unusual proteolysis within their transmembrane domains by unknown gamma-secretases. Here we show that an affinity reagent designed to interact with the active site of gamma-secretase binds directly and specifically to heterodimeric forms of presenilins, polytopic proteins that are mutated in hereditary Alzheimer's and are known mediators of gamma-secretase cleavage of both beta-APP and Notch. These results provide evidence that heterodimeric presenilins contain the active site of gamma-secretase, and validate presenilins as principal targets for the design of drugs to treat and prevent Alzheimer's disease.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Marcadores de Afinidad , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas , Células CHO , Cricetinae , Dimerización , Humanos , Proteínas de la Membrana/química , Microsomas/química , Microsomas/metabolismo , Peso Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Presenilina-1 , Presenilina-2 , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Unión Proteica , Procesamiento Proteico-Postraduccional , Transfección
8.
J Biol Chem ; 275(5): 3173-8, 2000 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-10652302

RESUMEN

The discovery that a deficiency of presenilin 1 (PS1) decreases the production of amyloid beta-protein (Abeta) identified the presenilins as important mediators of the gamma-secretase cleavage of beta-amyloid precursor protein (APP). Recently, we found that two conserved transmembrane (TM) aspartates in PS1 are critical for Abeta production, providing evidence that PS1 either functions as a required diaspartyl cofactor for gamma-secretase or is itself gamma-secretase. Presenilin 2 (PS2) shares substantial sequence and possibly functional homology with PS1. Here, we show that the two TM aspartates in PS2 are also critical for gamma-secretase activity, providing further evidence that PS2 is functionally homologous to PS1. Cells stably co-expressing TM Asp --> Ala mutations in both PS1 and PS2 show further accumulation of the APP-derived gamma-secretase substrates, C83 and C99. The production of Abeta is reduced to undetectable levels in the conditioned media of these cells. Furthermore, endoproteolysis of the exogenous Asp mutant PS2 is absent, and endogenous PS1 C-terminal fragments are diminished to undetectable levels. Therefore, the co-expression of PS1 and PS2 TM Asp --> Ala mutants suppresses the formation of any detectable PS1 or PS2 heterodimeric fragments and essentially abolishes the production of Abeta. These results explain the residual Abeta production seen in PS1-deficient cells and demonstrate the absolute requirement of functional presenilins for Abeta generation. We conclude that presenilins, and their TM aspartates in particular, are attractive targets for lowering Abeta therapeutically to prevent Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/química , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Células CHO , Cricetinae , Proteínas de la Membrana/metabolismo , Presenilina-1 , Presenilina-2
9.
Nature ; 398(6727): 513-7, 1999 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-10206644

RESUMEN

Accumulation of the amyloid-beta protein (Abeta) in the cerebral cortex is an early and invariant event in the pathogenesis of Alzheimer's disease. The final step in the generation of Abeta from the beta-amyloid precursor protein is an apparently intramembranous proteolysis by the elusive gamma-secretase(s). The most common cause of familial Alzheimer's disease is mutation of the genes encoding presenilins 1 and 2, which alters gamma-secretase activity to increase the production of the highly amyloidogenic Abeta42 isoform. Moreover, deletion of presenilin-1 in mice greatly reduces gamma-secretase activity, indicating that presenilin-1 mediates most of this proteolytic event. Here we report that mutation of either of two conserved transmembrane (TM) aspartate residues in presenilin-1, Asp 257 (in TM6) and Asp 385 (in TM7), substantially reduces Abeta production and increases the amounts of the carboxy-terminal fragments of beta-amyloid precursor protein that are the substrates of gamma-secretase. We observed these effects in three different cell lines as well as in cell-free microsomes. Either of the Asp --> Ala mutations also prevented the normal endoproteolysis of presenilin-1 in the TM6 --> TM7 cytoplasmic loop. In a functional presenilin-1 variant (carrying a deletion in exon 9) that is associated with familial Alzheimer's disease and which does not require this cleavage, the Asp 385 --> Ala mutation still inhibited gamma-secretase activity. Our results indicate that the two transmembrane aspartate residues are critical for both presenilin-1 endoproteolysis and gamma-secretase activity, and suggest that presenilin 1 is either a unique diaspartyl cofactor for gamma-secretase or is itself gamma-secretase, an autoactivated intramembranous aspartyl protease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ácido Aspártico/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Células CHO , Células COS , Línea Celular , Membrana Celular/metabolismo , Sistema Libre de Células , Coenzimas/metabolismo , Cricetinae , Electroquímica , Exones , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Microsomas/metabolismo , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/metabolismo , Presenilina-1 , Pliegue de Proteína , Proteínas Recombinantes/metabolismo , Transfección
10.
Biochemistry ; 37(47): 16465-71, 1998 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9843412

RESUMEN

Progressive cerebral deposition of the amyloid beta-protein (Abeta) is believed to play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The highly amyloidogenic 42-residue form of Abeta (Abeta42) is the first species to be deposited in both sporadic and familial AD. Mutations in two familial AD-linked genes, presenilins 1 (PS1) and 2 (PS2), selectively increase the production of Abeta42 in cultured cells and the brains of transgenic mice, and gene deletion of PS1 shows that it is required for normal gamma-secretase cleavage of the beta-amyloid precursor protein (APP) to generate Abeta. To establish the subcellular localization of the PS1 regulation of APP processing to Abeta, fibroblasts from PS1 wild-type (wt) or knockout (KO) embryos as well as Chinese hamster ovary (CHO) cells stably transfected with wt or mutant PS1 were subjected to subcellular fractionation on discontinuous Iodixanol gradients. APP C-terminal fragments (CTF) were markedly increased in both endoplasmic reticulum- (ER-) and Golgi-rich fractions of fibroblasts from KO mice; moreover, similar increases were documented directly in KO brain tissue. No change in the subcellular distribution of full-length APP was detectable in fibroblasts lacking PS1. In CHO cells, a small portion of APP, principally the N-glycosylated isoform, formed complexes with PS1 in both ER- and Golgi-rich fractions, as detected by coimmunoprecipitation. When the same fractions were analyzed by enzyme-linked immunosorbent assays for Abetatotal and Abeta42, Abeta42 was the major Abeta species in the ER fraction (Abeta42:Abetatotal ratio 0.5-1.0), whereas absolute levels of both Abeta42 and Abeta40 were higher in the Golgi fraction and the Abeta42:Abetatoal ratio was 0.05-0.16 there. Mutant PS1 significantly increased Abeta42 levels in the Golgi fraction. Our results indicate PS1 and APP can interact in the ER and Golgi, where PS1 is required for proper gamma-secretase processing of APP CTFs, and that PS1 mutations augment Abeta42 levels principally in Golgi-like vesicles.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/fisiología , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Células CHO , Células Cultivadas , Cricetinae , Retículo Endoplásmico/química , Retículo Endoplásmico/genética , Aparato de Golgi/química , Aparato de Golgi/genética , Líquido Intracelular/metabolismo , Riñón/citología , Sustancias Macromoleculares , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Presenilina-1 , Procesamiento Proteico-Postraduccional
11.
EMBO J ; 16(14): 4276-84, 1997 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-9250671

RESUMEN

The E1B 55-kDa and E4 34-kDa oncoproteins of adenovirus type 5 (abbreviated here as E1B-55kD and E4-34kD) promote the export of viral mRNA and inhibit the export of most cellular mRNA species. We show that the intracellular complex containing E1B-55kD and E4-34kD continuously shuttles between the nucleus and the cytoplasm, and may thus serve as a nucleocytoplasmic transporter for viral mRNA. We present evidence that within this complex, it is the E4-34kD protein that directs both nuclear import and nuclear export. E4-34kD contains a functional nuclear export signal similar to corresponding sequences found in the retroviral proteins rev and rex. This sequence element is required for nuclear export of the complex, and it can function autonomously when fused to a carrier protein and microinjected in HeLa cell nuclei. When E4-34kD is expressed alone, a portion of the protein that contains a predicted arginine-rich amphipathic alpha-helical structure mediates nuclear retention of the protein. This retention, however, can be abolished by the association with E1B-55kD or by a specific point mutation within the arginine-rich motif. The export of E4-34kD can be blocked by an HTLV-rex derived competitive inhibitor and overexpressed E4-34kD inhibits rev-mediated transport, suggesting that the export pathways accessed by the adenoviral and retroviral proteins share components. The interplay between two polypeptides as well as the involvement of a dominant nuclear retention domain are novel features that might contribute to the efficiency and regulation of the adenovirus export system.


Asunto(s)
Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Núcleo Celular/metabolismo , Señales de Clasificación de Proteína/metabolismo , Antígenos Transformadores de Poliomavirus/química , Antígenos Transformadores de Poliomavirus/genética , Proteínas Portadoras/metabolismo , Citoplasma/metabolismo , Productos del Gen rev/metabolismo , Productos del Gen rex/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Mutación/genética , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Transfección
12.
J Immunol ; 153(3): 1271-80, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8027554

RESUMEN

The presence of anti-neutrophil cytoplasmic Abs (ANCA) in many patients with systemic vasculitis suggests that ANCA may play a role in disease pathogenesis. Neutrophils from patients with Wegener's granulomatosis often express ANCA target Ags (myeloperoxidase (MPO) and proteinase 3 (PR3)) on their surface, making these intracellular primary granule enzymes accessible to these autoantibodies. Similarly, normal neutrophils can be induced to translocate MPO and PR3 to the cell surface in vitro, and we demonstrate that murine mAb ANCA IgG, but not IgM, binds to the ANCA target and engages the Fc gamma RIIa ligand-binding site on the surface of human neutrophils. In contrast to ANCA IgM, ANCA IgG also induces an oxidative burst in neutrophils (oxidation of dihydrorhodamine = 91 +/- 15 fluorescence units with anti-PR3 IgG vs 17 +/- 2 with anti-PR3 IgM, p < 0.001). Blockade of the ligand-binding site of Fc gamma RIIa with an antibinding site mAb Fab significantly reduces this ANCA IgG-triggered production of reactive oxygen species (p < 0.01). Similarly, human ANCA bind the ANCA target, engage Fc gamma RIIa, and induce an oxidative burst in neutrophils. The allelic phenotype of Fc gamma RIIa strongly influences the Fc gamma receptor engagement by ligand, and Fc gamma RIIa homozygous donors differ by more than threefold in the quantitative production of reactive oxygen intermediates (ROI) (p < 0.01). Thus, engagement of Fc gamma RIIa by the Fc region of ANCA is one mechanism by which these autoantibodies activate receptor-mediated signal transduction systems in human neutrophils to initiate programs of inflammation and tissue injury. Fc gamma receptor alleles may represent heritable disease risk factors influencing the magnitude of such a process.


Asunto(s)
Autoanticuerpos/inmunología , Neutrófilos/inmunología , Receptores de IgG/inmunología , Anticuerpos Anticitoplasma de Neutrófilos , Autoanticuerpos/metabolismo , Citoplasma/inmunología , Granulomatosis con Poliangitis/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Ligandos , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/metabolismo , Estallido Respiratorio , Vasculitis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...