Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 290(40): 24210-21, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26292220

RESUMEN

The single amino acid mutation G26R in human apolipoprotein A-I (apoA-I) is associated with familial amyloid polyneuropathy III. ApoA-I carrying this mutation (apoA-IIowa) forms amyloid fibrils in vitro. Heparan sulfate (HS) is a glycosaminoglycan that is abundant at the cell surface and in the extracellular matrix. Although HS and its highly sulfated domains are involved in aggregation of amyloid-ß and accumulate in cerebral amyloid plaques of patients with Alzheimer disease and mouse models of this disease, the role of HS in familial amyloid polyneuropathy III has never been addressed. Here, we used cell models to investigate the possible role of HS in the cytotoxicity of apoA-IIowa amyloid. Wild-type CHO cells, but not pgsD-677 cells, an HS-deficient CHO mutant, demonstrated uptake of apoA-IIowa amyloid after incubation with the amyloid. Addition of sulfated glycosaminoglycans to culture media prevented interaction with and cytotoxicity of apoA-IIowa amyloid to CHO cells. Elimination of cell surface HS or inhibition of HS sulfation with chemical reagents interfered with interaction of apoA-IIowa amyloid with CHO cells. We also found that cellular interaction and cytotoxicity of apoA-IIowa amyloid were significantly attenuated in CHO cells that stably expressed the human extracellular endoglucosamine 6-sulfatases HSulf-1 and HSulf-2. Our results thus suggest that cell surface HS mediates cytotoxicity of apoA-IIowa amyloid and that enzymatic remodeling of HS mitigates the cytotoxicity.


Asunto(s)
Neuropatías Amiloides Familiares/genética , Amiloide/química , Apolipoproteína A-I/química , Heparitina Sulfato/química , Sulfatos/química , Péptidos beta-Amiloides/química , Proteínas Amiloidogénicas/genética , Animales , Células CHO , Membrana Celular/química , Cricetinae , Cricetulus , Femenino , Glicósidos/química , Heparina/química , Humanos , Lisosomas/química , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , Unión Proteica , Estructura Terciaria de Proteína , Azufre/química , Porcinos
2.
J Biol Chem ; 290(34): 20947-20959, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26175149

RESUMEN

The N-terminal amino acid 1-83 fragment of apolipoprotein A-I (apoA-I) has a strong propensity to form amyloid fibrils at physiological neutral pH. Because apoA-I has an ability to bind to lipid membranes, we examined the effects of the lipid environment on fibril-forming properties of the N-terminal fragment of apoA-I variants. Thioflavin T fluorescence assay as well as fluorescence and transmission microscopies revealed that upon lipid binding, fibril formation by apoA-I 1-83 is strongly inhibited, whereas the G26R mutant still retains the ability to form fibrils. Such distinct effects of lipid binding on fibril formation were also observed for the amyloidogenic prone region-containing peptides, apoA-I 8-33 and 8-33/G26R. This amyloidogenic region shifts from random coil to α-helical structure upon lipid binding. The G26R mutation appears to prevent this helix transition because lower helical propensity and more solvent-exposed conformation of the G26R variant upon lipid binding were observed in the apoA-I 1-83 fragment and 8-33 peptide. With a partially α-helical conformation induced by the presence of 2,2,2-trifluoroethanol, fibril formation by apoA-I 1-83 was strongly inhibited, whereas the G26R variant can form amyloid fibrils. These findings suggest a new possible pathway for amyloid fibril formation by the N-terminal fragment of apoA-I variants: the amyloidogenic mutations partially destabilize the α-helical structure formed upon association with lipid membranes, resulting in physiologically relevant conformations that allow fibril formation.


Asunto(s)
Apolipoproteína A-I/química , Mutación , Fosfatidilcolinas/química , Proteínas Recombinantes de Fusión/química , Amiloide/química , Amiloide/genética , Apolipoproteína A-I/genética , Benzotiazoles , Escherichia coli/genética , Escherichia coli/metabolismo , Colorantes Fluorescentes , Expresión Génica , Humanos , Unión Proteica , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Relación Estructura-Actividad , Tiazoles , Trifluoroetanol/química , Liposomas Unilamelares/química
3.
Sci Rep ; 7(1): 2988, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28592796

RESUMEN

Apolipoprotein A-I (apoA-I) undergoes a large conformational reorganization during remodeling of high-density lipoprotein (HDL) particles. To detect structural transition of apoA-I upon HDL formation, we developed novel monoclonal antibodies (mAbs). Splenocytes from BALB/c mice immunized with a recombinant human apoA-I, with or without conjugation with keyhole limpet hemocyanin, were fused with P3/NS1/1-Ag4-1 myeloma cells. After the HAT-selection and cloning, we established nine hybridoma clones secreting anti-apoA-I mAbs in which four mAbs recognize epitopes on the N-terminal half of apoA-I while the other five mAbs recognize the central region. ELISA and bio-layer interferometry measurements demonstrated that mAbs whose epitopes are within residues 1-43 or 44-65 obviously discriminate discoidal and spherical reconstituted HDL particles despite their great reactivities to lipid-free apoA-I and plasma HDL, suggesting the possibility of these mAbs to detect structural transition of apoA-I on HDL. Importantly, a helix-disrupting mutation of W50R into residues 44-65 restored the immunoreactivity of mAbs whose epitope being within residues 44-65 against reconstituted HDL particles, indicating that these mAbs specifically recognize the epitope region in a random coil state. These results encourage us to develop mAbs targeting epitopes in the N-terminal residues of apoA-I as useful probes for monitoring formation and remodeling of HDL particles.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Lipoproteínas HDL/metabolismo , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Humanos , Ratones Endogámicos BALB C , Unión Proteica , Conformación Proteica
4.
Sci Rep ; 6: 30391, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27464946

RESUMEN

The single amino acid mutation G26R in human apolipoprotein A-I (apoA-IIowa) is the first mutation that was associated with familial AApoA1 amyloidosis. The N-terminal fragments (amino acid residues 1-83) of apoA-I containing this mutation deposit as amyloid fibrils in patients' tissues and organs, but the mechanisms of cellular degradation and cytotoxicity have not yet been clarified. In this study, we demonstrated degradation of apoA-IIowa fibrils via the autophagy-lysosomal pathway in human embryonic kidney 293 cells. ApoA-IIowa fibrils induced an increase in lysosomal pH and the cytosolic release of the toxic lysosomal protease cathepsin B. The mitochondrial dysfunction caused by apoA-IIowa fibrils depended on cathepsin B and was ameliorated by increasing the degradation of apoA-IIowa fibrils. Thus, although apoA-IIowa fibril transport to lysosomes and fibril degradation in lysosomes may have occurred, the presence of an excess number of apoA-IIowa fibrils, more than the lysosomes could degrade, may be detrimental to cells. Our results thus provide evidence that the target of apoA-IIowa fibrils is lysosomes, and we thereby gained a novel insight into the mechanism of AApoA1 amyloidosis.


Asunto(s)
Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Lisosomas/metabolismo , Actinas/metabolismo , Apolipoproteína A-I/química , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Línea Celular , Dinaminas/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Modelos Biológicos , Proteínas Mutantes , Agregado de Proteínas , Agregación Patológica de Proteínas , Proteolisis , Transducción de Señal
5.
FEBS Lett ; 590(20): 3492-3500, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27654470

RESUMEN

Glycosaminoglycans are known to be associated with extracellular amyloid deposits of various amyloidogenic proteins. In this study, we found that the glycosaminoglycan heparin greatly accelerates the elongation step in fibril formation by the N-terminal 1-83 fragment of human apolipoprotein A-I (apoA-I), especially in the amyloidogenic W50R variant. Using fragment peptides, we demonstrate that heparin significantly promotes ß-transition and fibril formation of the highly amyloidogenic region spanning residues 44-65 and colocalizes with fibrils formed by the W50R variant. These results suggest the possible role of glycosaminoglycans in fibril formation by amyloidogenic apoA-I variants.


Asunto(s)
Amiloide/metabolismo , Apolipoproteína A-I/genética , Heparina/metabolismo , Fragmentos de Péptidos/química , Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Dicroismo Circular , Humanos , Microscopía Electrónica de Transmisión , Mutación , Fragmentos de Péptidos/metabolismo , Estabilidad Proteica , Estructura Secundaria de Proteína , Relación Estructura-Actividad
6.
Amyloid ; 23(1): 17-25, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26701221

RESUMEN

INTRODUCTION: Apolipoprotein A-I (apoA-I) amyloidosis is either a non-hereditary form with deposits of wild-type apoA-I proteins in atherosclerotic plaques or a hereditary form with progressive accumulation of mutant apoA-I proteins in different tissues. Several small polyphenolic molecules reportedly inhibited formation of fibrillar assemblies of some amyloidogenic proteins and their cytotoxicity, but small molecules that inhibit apoA-I fibril formation have never been reported. METHODS: Our methods included a thioflavin-T-binding assay, atomic force microscopy and dot blot and cell-based assays. RESULTS: We showed that (-)-epigallocatechin-3-gallate (EGCG), a tea-derived flavanol, inhibited in vitro fibril formation and disaggregated fibrils preformed by the N-terminal 1-83 fragments of wild-type (WT) apoA-I and the G26R point mutation of apoA-I (apoA-IIowa). We eliminated a common structure recognized by the anti-amyloid antibody OC by incubating apoA-IIowa with EGCG or treating apoA-IIowa fibrils with EGCG, which supported the above observation. In addition, EGCG rescued human embryonic kidney 293 cells from cytotoxicity and attenuated production of reactive oxygen species, which were induced by apoA-IIowa fibrils. CONCLUSIONS: Our results support the concept that EGCG inhibits amyloid fibril formation of various amyloidogenic proteins. Thus, EGCG may be a candidate for providing a structure to develop de novo inhibitors for amyloidosis treatment.


Asunto(s)
Amiloide/metabolismo , Amiloidosis/prevención & control , Apolipoproteína A-I/metabolismo , Catequina/análogos & derivados , Catequina/farmacología , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Agregación Patológica de Proteínas/prevención & control , Estructura Secundaria de Proteína , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA