Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neurobiol Aging ; 38: 11-13, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26827638

RESUMO

There is a vast literature on the role of beta amyloid (Aß) peptides in the pathogenesis of Alzheimer's disease. However, there is a paucity of research on the potential physiological functions of these evolutionarily conserved products of the Aß precursor protein. Based on previous studies in neuroblastoma cells, we hypothesized that Aß may contribute to the proliferation of somatic cells. We present evidence supporting this hypothesis for the case of cultured human skin fibroblasts immortalized with the catalytic subunit of human telomerase (hTERT). Optimal concentrations ranged from 100 pM-10 nM, depending on the nature of the assay.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Proliferação de Células/genética , Fibroblastos/citologia , Fragmentos de Peptídeos/fisiologia , Linhagem Celular , Humanos , Pele/citologia , Telomerase
2.
Front Genet ; 4: 129, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23847654

RESUMO

Segmental progeroid syndromes are groups of disorders with multiple features suggestive of accelerated aging. One subset of adult-onset progeroid syndromes, referred to as atypical Werner syndrome, is caused by mutations in the LMNA gene, which encodes a class of nuclear intermediate filaments, lamin A/C. We previously described rapid telomere attrition and accelerated replicative senescence in cultured fibroblasts overexpressing mutant lamin A. In this study, we investigated the cellular phenotypes associated with accelerated telomere shortening in LMNA mutant primary fibroblasts. In early passage primary fibroblasts with R133L or L140R LMNA mutations, shelterin protein components were already reduced while cells still retained telomere lengths comparable to those of controls. There was a significant inverse correlation between the degree of abnormal nuclear morphology and the level of TRF2, a shelterin subunit, suggesting a potential causal relationship. Stabilization of the telomeres via the introduction of the catalytic subunit of human telomerase, hTERT (human telomerase reverse transcriptase), did not prevent degradation of shelterin components, indicating that reduced TRF2 in LMNA mutants is not mediated by short telomeres. Interestingly, γ-H2AX foci (reflecting double strand DNA damage) in early passage LMNA mutant primary fibroblasts and LMNA mutant hTERT fibroblasts were markedly increased in non-telomeric regions of DNA. Our results raise the possibility that mutant lamin A/C causes global genomic instability with accumulation of non-telomeric DNA damage as an early event, followed by TRF2 degradation and telomere shortening.

3.
Mol Vis ; 17: 2672-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22065920

RESUMO

PURPOSE: To determine the differences between species in the retention of lens fiber cell nuclei and nuclear fragments in the aging lens cortex and the relationship of nuclear retention to lens opacity. For this purpose old human, monkey, dog, and rat lenses were compared to those of three strains of mouse. We also investigated possible mechanisms leading to nuclear retention. METHODS: Fixed specimens of the species referred to above were obtained from immediate on site sacrifice of mice and rats, or from recently fixed lenses of other species, dogs, monkeys, and humans, obtained from collaborators. The retention of undegraded nuclei and nuclear fragments was graded 1-4 from histologic observation. All species lenses were examined microscopically in fixed sections stained with hematoxylin and eosin (H&E) or 4',6-diamidino-2-phenylindole (DAPI). Slit lamp observations were made only on the mice and rats before sacrifice and lens fixation. Values of 0 to 4 (clear lens to cataract) were given to degree of opacity. MRNA content in young versus old C57BL/6 mouse lenses was determined by quantitative PCR (qPCR) for DNase II-like acid DNase (DLAD) and other proteins. DLAD protein was determined by immunofluorescence of fixed eye sections. RESULTS: In old C57BL/6 and DBA mice and, to a lesser degree, in old CBA mice and old Brown Norway (BN) rats lenses were seen to contain a greatly expanded pool of unresolved whole nuclei or fragments of nuclei in differentiating lens fiber cells. This generally correlated with increased slit lamp opacities in these mice. Most old dog lenses also had an increase in retained cortical nuclei, as did a few old humans. However, a second rat strain, BNF1, in which opacity was quite high had no increase in retained nuclei with age nor did any of the old monkeys, indicating that retained nuclei could not be a cause of opacity in these animals. The nuclei and nuclear fragments were located at all levels in the outer cortex extending inward from the lens equator and were observable by the DAPI. These nuclei and nuclear fragments were seen from 12 months onward in all C57BL/6 and DBA/2 mice and to a lesser degree in the CBA, increasing in number and in space occupancy with increasing age. Preliminary results suggest that retention of nuclei in the C57BL/6 mouse is correlated with an age-related loss of DLAD from old lenses. CONCLUSIONS: A very marked apparently light refractive condition caused by retained cortical nuclei and nuclear fragments is present in the lens cortices, increasing with age in the three strains of mice examined and in one of two strains of rats (BN). This condition was also seen in some old dogs and a few old humans. It may be caused by an age-related loss of DLAD, which is essential for nuclear DNA degradation in the lens. However, this condition does not develop in old BNF1 rats, or old monkeys and is only seen sporadically in humans. Thus, it can not be a universal cause for age related lens opacity or cataract presence, although it develops concurrently with opacity in mice. This phenomenon should be considered when using the old mouse as a model for human age-related cataract.


Assuntos
Envelhecimento , Catarata/patologia , Núcleo Celular/patologia , Endodesoxirribonucleases/deficiência , Fibrose/patologia , Córtex do Cristalino/patologia , Idoso , Idoso de 80 Anos ou mais , Animais , Catarata/complicações , Catarata/metabolismo , Núcleo Celular/metabolismo , DNA/metabolismo , Fragmentação do DNA , Cães , Endodesoxirribonucleases/genética , Feminino , Fibrose/complicações , Fibrose/metabolismo , Haplorrinos , Histocitoquímica , Humanos , Indóis/análise , Córtex do Cristalino/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos BN , Projetos de Pesquisa , Especificidade da Espécie
4.
Mol Vis ; 16: 1496-513, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20806081

RESUMO

PURPOSE: To compare age-related cataractous (ARC) changes in unirradiated mice lenses to those induced by head-only X-irradiation of 3 month-old mice. METHODS: lens epithelial cells (LECs) as well as partially degraded cortical DNA were visualized in fixed sections using 4',6-diamidino-2-phenylindole (DAPI) staining, and in fresh lenses using the vital stain Hoechst 33342. reactive oxygen species (ROS) activity was also visualized directly in fresh lenses using the vital dye Dihydrorhodamine (DHR). In fixed lenses an antibody specific for 8-OH Guanosine (8-OH-G) lesions was used to visualize DNA oxidative adducts from ROS damage. Alpha smooth muscle actin was visualized using specific antibodies to determine if myofibroblasts were present. Fluorescence was quantified using Laser Scanning Confocal Microscopy (LSCM). The degree of lens opacity and cataract formation was determined by slit lamp, or from digitalized images of light reflections taken with a low magnification light microscope. RESULTS: Using DNA- and ROS-specific vital fluorescent dyes, and laser scanning confocal microscopy we have previously described 4 changes in the aging rodent lenses: 1) a significantly decreased density of surface LECs in lenses from old compared to younger mice and rats; 2) a very large increase in retained cortical nuclei and DNA fragments in the secondary lens fibers of old rodent lenses; 3) increased cortical ROS in old rodent lenses; 4) increased cataract concomitantly with the cortical DNA and ROS increases. In the current study we report that these same 4 changes also occur in an accelerated fashion in mice given head-only X-irradiation at 3 months of age. In addition to vital staining of fresh lenses, we also examined sections from fixed eyes stained with DAPI or hematoxylin and eosin (H&E) and found the same loss of surface LECs and accumulation of undigested nuclei and debris in secondary lens fibers occur with age or following X-irradiation. In addition sections from fixed-eyes were examined for ROS damage to DNA with antibodies specific for 8-OH-G lesions. The frequency of 8-OH-G lesions increased dramatically in lenses from old unirradiated mice over 24 months of age, and similarly in X-irradiated lenses by 9-11 months post irradiation. The accumulation of cortical nuclei was not the result of conversion or invasion by myofibroblasts as tested by antibodies to a marker for such cells, alpha smooth muscle actin. CONCLUSIONS: X-irradiation damage induces a large decrease in surface LECs over a period of 3-11 months post X-irradiation of young mice. These changes are similar in extent to those seen in 24-29 months-old control mouse lenses with age-related cataracts. In 24+ month-old unirradiated mice the secondary lens fibers are not able to degrade nuclei or nuclear DNA efficiently and accumulate large numbers of cortical nuclei and nuclear fragments as well as ROS and 8-OHG lesions. X-irradiated lenses develop the same abnormalities in a more accelerated fashion. The extensive loss of LECS and accumulation of undegraded nuclei, ROS, and ROS damage may play a causal role in cataract generation in both unirradiated old mice and in previously irradiated young adult mice.


Assuntos
Envelhecimento/patologia , Catarata/patologia , DNA/metabolismo , Células Epiteliais/patologia , Córtex do Cristalino/metabolismo , Córtex do Cristalino/patologia , Espécies Reativas de Oxigênio/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Animais , Catarata/induzido quimicamente , Catarata/metabolismo , Contagem de Células , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Células Epiteliais/metabolismo , Feminino , Indóis/metabolismo , Córtex do Cristalino/cirurgia , Núcleo do Cristalino/metabolismo , Núcleo do Cristalino/patologia , Camundongos , Camundongos Endogâmicos C57BL , Coloração e Rotulagem , Fixação de Tecidos , Raios X
5.
J Neurochem ; 112(2): 410-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19860855

RESUMO

FE65 is a multi-modular adaptor protein that binds the cytoplasmic tail of the beta-amyloid precursor protein (APP). Genetic evidence suggests that APP is intimately involved in the pathogenesis of dementias of the Alzheimer type, neurodegenerative disorders that affect multiple cognitive domains, including learning and memory. Evidence from p97FE65-specific knockout mice (lacking the 97 kDa full-length FE65 protein, p97FE65) suggests an important role for FE65 in learning and memory. Interpretation of the learning and memory phenotype, however, is complicated by the up-regulation (compared with wild-type mice) of a novel 60 kDa FE65 isoform (p60FE65). Here, we report an evidence that p60FE65 is translated from an alternative methionine, M261, on the p97FE65 transcript. Thus, p60FE65 has a shortened N-terminus, lacking part of the WW domain that is considered important for nuclear translocation and transactivation of gene expression. Consistently, p60FE65 exhibits an attenuated ability for APP-Gal4-mediated transcription as compared with p97FE65. Similar to p97FE65, however, both transfected and endogenous p60FE65 are able to translocate to the nucleus in cultured cells and in neurons. These results are consistent with earlier evidence from our laboratory that reduced FE65 nuclear signaling may contribute, in part, to the phenotypes observed in p97FE65 knockout mice.


Assuntos
Transtornos Cognitivos/fisiopatologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Regulação para Cima/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Humanos , Indóis , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Peso Molecular , Neuroblastoma , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/genética , Transporte Proteico/genética , Transdução de Sinais/genética , Transfecção/métodos
6.
J Mol Neurosci ; 31(1): 59-68, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17416970

RESUMO

Dementia of the Alzheimer type (DAT) is associated with the accumulation of beta-amyloid (A beta) peptides derived from beta-amyloid precursor protein (APP). Goldstein and coworkers have suggested that APP acts as a cargo receptor connecting post-Golgi vesicles and motor proteins. Sisodia and colleagues have suggested that APP is a passive passenger within the vesicles. Both views predict that one should be able to visualize colocalizations of APP with microtubules, the object of the present investigation. To avoid possible artifacts created by APP overexpression, we studied endogenous expression in a human neuroblastoma cell line (SK-N-SH). Using high resolution fluorescence microscopy and antibodies specific for the amino termini of APP and A beta sequences, we found that endogenous APP and A beta peptide immunoreactivities colocalized with microtubules in interphase cells. Disruption of microtubules, followed by fixation at various time points during repolymerization, allowed us to observe the sequence and timing of these colocalizations in interphase cells. In addition, to our surprise, we found that A beta immunoreactivities colocalize with the mitotic spindle, a bundle of specialized microtubules. Because of the condensed cytoplasm found in neurons, we suggest that SK-N-SH cells might be a more convenient experimental system for exploring the mechanisms that underlie these protein localizations and the pathology that might result from altered APP protein structure and function.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Microtúbulos/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/metabolismo , Animais , Transporte Biológico/fisiologia , Linhagem Celular Tumoral , Epitopos , Humanos , Imuno-Histoquímica , Interfase , Modelos Biológicos , Neuroblastoma , Fuso Acromático
7.
J Biol Chem ; 280(13): 12548-58, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15647266

RESUMO

Adaptor protein FE65 (APBB1) specifically binds to the intracellular tail of the type I transmembrane protein, beta-amyloid precursor protein (APP). The formation of this complex may be important for modulation of the processing and function of APP. APP is proteolytically cleaved at multiple sites. The cleavages and their regulation are of central importance in the pathogenesis of dementias of the Alzheimer type. In cell cultures and perhaps in vivo, secretion of the alpha-cleaved APP ectodomain (sAPPalpha) is the major pathway in the most cells. Regulation of the process may require extracellular/intracellular cues. Neither extracellular ligands nor intracellular mediators have been identified, however. Here, we show novel evidence that the major isoform of FE65 (97-kDa FE65, p97FE65) can be converted to a 65-kDa N-terminally truncated C-terminal fragment (p65FE65) via endoproteolysis. The cleavage region locates immediately after an acidic residue cluster but before the three major protein-protein binding domains. The cleavage activity is particularly high in human and non-human primate cells and low in rodent cells; the activity appears to be triggered/enhanced by high cell density, presumably via cell-cell/cell-substrate contact cues. As a result, p65FE65 exhibits extraordinarily high affinity for APP (up to 40-fold higher than p97FE65) and potent suppression (up to 90%) of secretion of sAPPalpha. Strong p65FE65-APP binding is required for the suppression. The results suggest that p65FE65 may be an intracellular mediator in a signaling cascade regulating alpha-secretion of APP, particularly in primates.


Assuntos
Precursor de Proteína beta-Amiloide/química , Proteínas do Tecido Nervoso/química , Proteínas Nucleares/química , Idoso , Sequência de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Encéfalo/metabolismo , Células COS , Feminino , Glutationa Transferase/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA